instruction
stringlengths
34
186
input
stringlengths
2.02k
93.8k
output
stringlengths
2
418
meta_questiontype
stringclasses
6 values
meta_inputlicense
stringclasses
6 values
meta_pmid
stringlengths
8
8
meta_safetyreportid
int64
9.51M
21M
meta_articlepubdate
stringlengths
4
10
What was the dosage of drug 'DONEPEZIL HYDROCHLORIDE'?
Development of donepezil-induced hypokalemia following treatment of cognitive impairment. Donepezil is a cholinesterase inhibitor used extensively to treat Alzheimer disease. The increased cholinergic activity is associated with adverse effects, therefore gastrointestinal symptoms, including nausea, vomiting, and diarrhea, are common. Hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients. Although hypokalemia of mild and moderate grade does not present serious signs and symptoms, severe hypokalemia often results in prolonged hospitalization and mortality. Herein, we report a case of hypokalemia developed after the initiation of donepezil therapy for cognitive impairment. Introduction Hypokalemia is a common electrolyte disturbance in clinical practice. The major causes of hypokalemia include gastrointestinal loss and medications such as diuretics [1,2]. Most cases are asymptomatic and mild, but some patients develop severe hypokalemia resulting in arrhythmias and patient death. About 20% of inpatients experience hypokalemia during hospitalization [3], and among these cases, severe hypokalemia was sometimes associated with prolonged hospitalization and increased mortality [4]. Donepezil is the second approved acetylcholinesterase inhibitor for the treatment of mild to moderate Alzheimer disease by the United States Food and Drug Administration (FDA), extensively used worldwide [5]. As donepezil is generally tolerated, most adverse events are gastrointestinal symptoms, including vomiting and diarrhea [6]. However, rare adverse events such as lupus, psychosis, and arrhythmia have been reported in a few patients [7,8]. According to the FDA, hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients [9,10]. Notably, hypokalemia exhibits non-specific symptoms such as general weakness, fatigue, dyspepsia, myalgia, tingling sensation, muscle cramps, and spasms; hence, it can be difficult to diagnose without laboratory investigations. Therefore, clinicians sometimes fail to recognize the signs and symptoms of hypokalemia. Accordingly, a rare case of hypokalemia induced by medication is significant. Herein, we report a case of hypokalemia developed after initiating donepezil as a treatment for cognitive impairment. Case The study was approved by the Institutional Review Board of the Catholic University of Korea (IRB No: PC20ZASI0046) with waiver of informed consent. An 87-year-old man visited the outpatient clinic owing to poor oral intake. According to his medical history, he had undergone surgery for benign prostatic hyperplasia 6 years ago and had discontinued the medication for benign prostatic hyperplasia 4 months before hospitalization. Recently, he was diagnosed with cognitive dysfunction based on a mini-mental status examination score of 22, and a global deterioration scale of 3. And was started on appropriate medication. At the time of admission, he was prescribed choline alfoscerate 400 mg twice daily and donepezil 5 mg for the control of Alzheimer disease. A review of systems did not reveal abnormal findings. His vital signs were as follows: blood pressure (BP), 180/70 mmHg; pulse rate, 72 beats/min; respiratory rate, 20 breaths/min; body temperate, 36.7℃. To determine the cause of poor oral intake, routine investigations were performed, including complete blood count, blood chemistry, urine analysis, electrocardiogram, and chest X-ray. The presence of subtle pneumonia on the chest X-ray and hypokalemia (serum potassium 2.6 mmol/L) was detected. Initial serum inflammatory markers showed the following results: plasma leukocyte count, 7,500/mm3; serum c-reactive protein, 0.74 mg/dL (range, 0.01–0.5 mg/dL). Except for potassium, blood chemistry revealed the following: serum albumin, 4.0 g/dL (range, 3.5–5.2 g/dL); blood urea nitrogen, 10.9 mg/dL; serum creatinine, 0.72 mg/dL (range, 0.61–1.20 mg/dL); serum sodium, 140 mmol/L; serum chloride, 91 mmol/L; serum magnesium, 2.4 mg/dL; serum osmolality, 290 mOsm/kg. Urinalysis and urine sediment examination presented the following: urine pH, 7.5; urine specific gravity, 1.011; urine white blood cells, 0–2/high power field; urine red blood cells, 0–2/high power field. Blood gas analysis revealed compensated metabolic alkalosis as follows: arterial blood pH, 7.52; arterial blood pCO2, 46.6 mmHg; arterial blood pO2, 73.7 mmHg; arterial blood HCO3¯, 36.8 mmol/L. Chest X-ray and inflammatory markers suggested that the pneumonia was not severe. Therefore, hypokalemia could be the cause of underlying poor oral intake. The patient was admitted to treat pneumonia and hypokalemia. For pneumonia treatment, he received ceftriaxone and clarithromycin as antibiotic agents. To determine the cause of hypokalemia, further investigations were performed. No clues indicating hypokalemia were detected in the medical history and review of systems. Next, his renal potassium excretion was measured to determine the potassium/creatinine ratio, fractional excretion of potassium, and transtubular potassium gradient (TTKG), using blood chemistry and spot urine chemistry. The results of spot urine chemistry were as follows: urea nitrogen, 392.8 mg/dL; creatinine, 90.8 mg/dL; total protein, 29.9 mg/dL; sodium, 35 mEq/L; potassium, 29.2 mEq/L; chloride, 45 mEq/L; osmolality, 309 mOsm/kg. Additionally, the following values were determined and suggested renal potassium wasting: urine potassium/creatinine ratio, 32.2 mEq/g; fractional excretion of potassium, 8.9%; TTKG, 10.5. Those findings suggested renal potassium wasting. Next, computed tomography demonstrated no abnormalities in the bilateral adrenal glands and both kidneys (Fig. 1). A hormone study was conducted to evaluate the cause of high BP. The thyroid function test was in the normal range. The adrenocorticotropic hormone (ACTH) level was 36.02 pg/mL (range, 10.0–60.0 pg/mL) and the cortisol level was 10.51 µg/dL (range, 9.41–26.06 µg/dL), which were within the normal range, revealing no mineral corticoid excess. Plasma renin activity was determined as 0.19 ng/mL/hr (range, 0.3–2.9 ng/mL/hr), aldosterone concentration was 14.93 pg/mL (range, 29.9–158.8 pg/mL), and the aldosterone/renin ratio was 7.86, indicating hyporeninemic hypoaldosteronism. However, unlike Liddle syndrome, the serum sodium level was normal at 140 mmol/L. The serum magnesium level was normal at 2.4 mg/dL, which was far from that observed in Gitelman syndrome. Hence, no obvious cause of hypokalemia was determined. During investigations analyzing the possible cause of hypokalemia, he underwent potassium supplementation using intravenous and oral formulations. Until the fourth day of hospitalization, his serum potassium level demonstrated difficulty increasing beyond 3.0 mmol/L despite sufficient potassium administration (Fig. 2). As the hypokalemia was poorly corrected, clinicians suspected other causes of hypokalemia. A thorough review of the therapeutic agents prescribed for cognitive dysfunction was conducted. On examining possible adverse effects, we identified evidence suggesting that donepezil causes hypokalemia as a rare adverse event [9,10]. Therefore, donepezil was immediately discontinued. Thereafter, his potassium demand, supplemented intravenously, gradually decreased and was modified to oral potassium agents. On the third day after donepezil cessation, his serum potassium level recovered to 3.5 mmol/L under oral potassium supplementation of 32 mEq/day (Fig. 2). During hypokalemia evaluation and correction, pneumonia was properly controlled. Finally, he was able to maintain serum potassium of 3.3 mmol/L under oral potassium supplementation of 32 mEq/day and was discharged on the ninth day of hospitalization. At the time of discharge, the results of blood chemistry and spot urine chemistry were as follows: serum creatinine, 0.73 mg/dL; serum potassium, 3.5 mmol/L; serum magnesium, 2.3 mg/dL; serum osmolality, 295 mOsm/kg; urine creatinine, 120.6 mg/dL; potassium, 46.7 mEq/L; osmolality, 330 mOsm/kg. Additionally, the following values were determined: urine potassium/creatinine ratio, 38.7 mEq/g; fractional excretion of potassium, 8.1%; TTKG, 11.9. Conversely, in the past, he had presented a systolic BP of less than 130 mmHg and diastolic BP of 80 mmHg during several visits to the outpatient clinic, with no history of medication-related hypertension. However, for 24 hours after hospitalization, BP was continuously confirmed as 160/90 mmHg or more, including a maximum of 185/105 mmHg, and amlodipine 5 mg was initiated. As BP measurements were above 140/90 mmHg on average after the addition of amlodipine 5 mg, the patient was additionally prescribed olmesartan 20 mg on the fifth day of hospitalization. Finally, in the outpatient clinic follow-ups, serum potassium was measured as 4.5 mmol/L, and oral potassium agents were withheld. Discussion This case report presents the rare development of hypokalemia with donepezil, an agent used to treat cognitive dysfunction. The most common cause of hypokalemia is gastrointestinal losses, followed by medications such as diuretics [1,2]. Additionally, various conditions lead to hypokalemia. For example, the following factors result in the development of renal potassium wasting: malignant hypertension, renal artery stenosis, renin-secreting tumors that can increase renin, adrenal hyperplasia, Cushing syndrome, medication including diuretics, magnesium deficiency, Gitelman syndrome, and chronic metabolic acidosis [3]. However, it is difficult to implicate donepezil, used for cognitive dysfunction therapy, as a causative agent. If this patient had multiple prescriptions, donepezil was probably not considered a major cause of hypokalemia. Donepezil is a cholinesterase inhibitor, mainly prescribed for Alzheimer disease. By inhibiting acetylcholinesterase, donepezil improves behavioral and cognitive symptoms, including confusion, aggression, and psychosis [11,12]. In several studies, donepezil has demonstrated improved cognitive functions in patients with dementia, but it had some adverse effects [13]. The adverse effects were associated with increased cholinergic activity, and the gastrointestinal system was mainly affected. Therefore, nausea, vomiting, and diarrhea were the most common symptoms, as well as insomnia, abnormal dreams, hepatotoxicity, and cardiovascular adverse events [14]. Hypokalemia was one of the rare adverse effects. The mechanism by which donepezil causes hypokalemia remains unclear. In this case, based on TTKG, potassium excretion continued under donepezil therapy. Therefore, we hypothesized the mechanism by which donepezil caused renal potassium wasting. This finding may be due to the action of donepezil on the ion channels in the renal tubule or Henle's loop. Donepezil could potentially inhibit the Na+-K+-2Cl- cotransporter in the thick ascending limb of Henle's loop, suppressing potassium reabsorption. Additionally, it could be postulated that donepezil stimulates the renal outer medullary potassium channel to excrete potassium. As another hypothesis, donepezil could affect the sodium channel epithelium in the principal cell of the collecting tubule. Thereafter, potassium appeared to be secreted into the lumen to maintain electrolyte balance. Although the mechanism by which donepezil induces hypokalemia remains unclear, we anticipated that renal potassium wasting would recover after its withdrawal. Unfortunately, the follow-up study did not achieve demonstrate an improvement in renal potassium wasting. Based on previous reports, we speculated the possible reasons by which recovery from renal potassium wasting may vary from 2 days to 3 months [15,16]. Therefore, the patient may require further time for recovery. Nonetheless, this case was significant as it confirmed the progression of hypokalemia recovery, necessitating a decrease in the daily amount of potassium supplementation after the withdrawal of donepezil. A limitation of this case report is that we have not done additional research regarding how donepezil affects the renal ion channels. Further case reports and research would assist in determining the mechanism by which donepezil causes renal potassium wasting. Although donepezil demonstrates rare serious adverse events, symptoms during early therapy require a differential diagnosis for hypokalemia. Conflicts of interest No potential conflict of interest relevant to this article was reported. Author contributions Conceptualization: all authors; Data curation: DK, THB, HSP; Formal analysis: DK, THB, HEY, BSC, BSK; Investigation: DK, THB, HEY, HSP; Methodology, Project administration: THB; Supervision: THB, BSC, BSK; Validation: THB, SJS, BSC, BSK; Writing-original draft: DK; Writing-review & editing: DK, THB, HEY, HSP, SJS. Fig. 1. Abdominal computed tomography scan. (A) Both adrenal glands (arrows) show no abnormalities. (B) Both kidneys (arrows) show no abnormal findings. Fig. 2. Summary of serum potassium level, potassium supplementation, total intake, and total output per day. IV, intravenous; PO, per oral.
5 mg (milligrams).
DrugDosage
CC BY-NC
33045804
18,435,143
2021-01
What was the dosage of drug 'DONEPEZIL'?
Development of donepezil-induced hypokalemia following treatment of cognitive impairment. Donepezil is a cholinesterase inhibitor used extensively to treat Alzheimer disease. The increased cholinergic activity is associated with adverse effects, therefore gastrointestinal symptoms, including nausea, vomiting, and diarrhea, are common. Hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients. Although hypokalemia of mild and moderate grade does not present serious signs and symptoms, severe hypokalemia often results in prolonged hospitalization and mortality. Herein, we report a case of hypokalemia developed after the initiation of donepezil therapy for cognitive impairment. Introduction Hypokalemia is a common electrolyte disturbance in clinical practice. The major causes of hypokalemia include gastrointestinal loss and medications such as diuretics [1,2]. Most cases are asymptomatic and mild, but some patients develop severe hypokalemia resulting in arrhythmias and patient death. About 20% of inpatients experience hypokalemia during hospitalization [3], and among these cases, severe hypokalemia was sometimes associated with prolonged hospitalization and increased mortality [4]. Donepezil is the second approved acetylcholinesterase inhibitor for the treatment of mild to moderate Alzheimer disease by the United States Food and Drug Administration (FDA), extensively used worldwide [5]. As donepezil is generally tolerated, most adverse events are gastrointestinal symptoms, including vomiting and diarrhea [6]. However, rare adverse events such as lupus, psychosis, and arrhythmia have been reported in a few patients [7,8]. According to the FDA, hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients [9,10]. Notably, hypokalemia exhibits non-specific symptoms such as general weakness, fatigue, dyspepsia, myalgia, tingling sensation, muscle cramps, and spasms; hence, it can be difficult to diagnose without laboratory investigations. Therefore, clinicians sometimes fail to recognize the signs and symptoms of hypokalemia. Accordingly, a rare case of hypokalemia induced by medication is significant. Herein, we report a case of hypokalemia developed after initiating donepezil as a treatment for cognitive impairment. Case The study was approved by the Institutional Review Board of the Catholic University of Korea (IRB No: PC20ZASI0046) with waiver of informed consent. An 87-year-old man visited the outpatient clinic owing to poor oral intake. According to his medical history, he had undergone surgery for benign prostatic hyperplasia 6 years ago and had discontinued the medication for benign prostatic hyperplasia 4 months before hospitalization. Recently, he was diagnosed with cognitive dysfunction based on a mini-mental status examination score of 22, and a global deterioration scale of 3. And was started on appropriate medication. At the time of admission, he was prescribed choline alfoscerate 400 mg twice daily and donepezil 5 mg for the control of Alzheimer disease. A review of systems did not reveal abnormal findings. His vital signs were as follows: blood pressure (BP), 180/70 mmHg; pulse rate, 72 beats/min; respiratory rate, 20 breaths/min; body temperate, 36.7℃. To determine the cause of poor oral intake, routine investigations were performed, including complete blood count, blood chemistry, urine analysis, electrocardiogram, and chest X-ray. The presence of subtle pneumonia on the chest X-ray and hypokalemia (serum potassium 2.6 mmol/L) was detected. Initial serum inflammatory markers showed the following results: plasma leukocyte count, 7,500/mm3; serum c-reactive protein, 0.74 mg/dL (range, 0.01–0.5 mg/dL). Except for potassium, blood chemistry revealed the following: serum albumin, 4.0 g/dL (range, 3.5–5.2 g/dL); blood urea nitrogen, 10.9 mg/dL; serum creatinine, 0.72 mg/dL (range, 0.61–1.20 mg/dL); serum sodium, 140 mmol/L; serum chloride, 91 mmol/L; serum magnesium, 2.4 mg/dL; serum osmolality, 290 mOsm/kg. Urinalysis and urine sediment examination presented the following: urine pH, 7.5; urine specific gravity, 1.011; urine white blood cells, 0–2/high power field; urine red blood cells, 0–2/high power field. Blood gas analysis revealed compensated metabolic alkalosis as follows: arterial blood pH, 7.52; arterial blood pCO2, 46.6 mmHg; arterial blood pO2, 73.7 mmHg; arterial blood HCO3¯, 36.8 mmol/L. Chest X-ray and inflammatory markers suggested that the pneumonia was not severe. Therefore, hypokalemia could be the cause of underlying poor oral intake. The patient was admitted to treat pneumonia and hypokalemia. For pneumonia treatment, he received ceftriaxone and clarithromycin as antibiotic agents. To determine the cause of hypokalemia, further investigations were performed. No clues indicating hypokalemia were detected in the medical history and review of systems. Next, his renal potassium excretion was measured to determine the potassium/creatinine ratio, fractional excretion of potassium, and transtubular potassium gradient (TTKG), using blood chemistry and spot urine chemistry. The results of spot urine chemistry were as follows: urea nitrogen, 392.8 mg/dL; creatinine, 90.8 mg/dL; total protein, 29.9 mg/dL; sodium, 35 mEq/L; potassium, 29.2 mEq/L; chloride, 45 mEq/L; osmolality, 309 mOsm/kg. Additionally, the following values were determined and suggested renal potassium wasting: urine potassium/creatinine ratio, 32.2 mEq/g; fractional excretion of potassium, 8.9%; TTKG, 10.5. Those findings suggested renal potassium wasting. Next, computed tomography demonstrated no abnormalities in the bilateral adrenal glands and both kidneys (Fig. 1). A hormone study was conducted to evaluate the cause of high BP. The thyroid function test was in the normal range. The adrenocorticotropic hormone (ACTH) level was 36.02 pg/mL (range, 10.0–60.0 pg/mL) and the cortisol level was 10.51 µg/dL (range, 9.41–26.06 µg/dL), which were within the normal range, revealing no mineral corticoid excess. Plasma renin activity was determined as 0.19 ng/mL/hr (range, 0.3–2.9 ng/mL/hr), aldosterone concentration was 14.93 pg/mL (range, 29.9–158.8 pg/mL), and the aldosterone/renin ratio was 7.86, indicating hyporeninemic hypoaldosteronism. However, unlike Liddle syndrome, the serum sodium level was normal at 140 mmol/L. The serum magnesium level was normal at 2.4 mg/dL, which was far from that observed in Gitelman syndrome. Hence, no obvious cause of hypokalemia was determined. During investigations analyzing the possible cause of hypokalemia, he underwent potassium supplementation using intravenous and oral formulations. Until the fourth day of hospitalization, his serum potassium level demonstrated difficulty increasing beyond 3.0 mmol/L despite sufficient potassium administration (Fig. 2). As the hypokalemia was poorly corrected, clinicians suspected other causes of hypokalemia. A thorough review of the therapeutic agents prescribed for cognitive dysfunction was conducted. On examining possible adverse effects, we identified evidence suggesting that donepezil causes hypokalemia as a rare adverse event [9,10]. Therefore, donepezil was immediately discontinued. Thereafter, his potassium demand, supplemented intravenously, gradually decreased and was modified to oral potassium agents. On the third day after donepezil cessation, his serum potassium level recovered to 3.5 mmol/L under oral potassium supplementation of 32 mEq/day (Fig. 2). During hypokalemia evaluation and correction, pneumonia was properly controlled. Finally, he was able to maintain serum potassium of 3.3 mmol/L under oral potassium supplementation of 32 mEq/day and was discharged on the ninth day of hospitalization. At the time of discharge, the results of blood chemistry and spot urine chemistry were as follows: serum creatinine, 0.73 mg/dL; serum potassium, 3.5 mmol/L; serum magnesium, 2.3 mg/dL; serum osmolality, 295 mOsm/kg; urine creatinine, 120.6 mg/dL; potassium, 46.7 mEq/L; osmolality, 330 mOsm/kg. Additionally, the following values were determined: urine potassium/creatinine ratio, 38.7 mEq/g; fractional excretion of potassium, 8.1%; TTKG, 11.9. Conversely, in the past, he had presented a systolic BP of less than 130 mmHg and diastolic BP of 80 mmHg during several visits to the outpatient clinic, with no history of medication-related hypertension. However, for 24 hours after hospitalization, BP was continuously confirmed as 160/90 mmHg or more, including a maximum of 185/105 mmHg, and amlodipine 5 mg was initiated. As BP measurements were above 140/90 mmHg on average after the addition of amlodipine 5 mg, the patient was additionally prescribed olmesartan 20 mg on the fifth day of hospitalization. Finally, in the outpatient clinic follow-ups, serum potassium was measured as 4.5 mmol/L, and oral potassium agents were withheld. Discussion This case report presents the rare development of hypokalemia with donepezil, an agent used to treat cognitive dysfunction. The most common cause of hypokalemia is gastrointestinal losses, followed by medications such as diuretics [1,2]. Additionally, various conditions lead to hypokalemia. For example, the following factors result in the development of renal potassium wasting: malignant hypertension, renal artery stenosis, renin-secreting tumors that can increase renin, adrenal hyperplasia, Cushing syndrome, medication including diuretics, magnesium deficiency, Gitelman syndrome, and chronic metabolic acidosis [3]. However, it is difficult to implicate donepezil, used for cognitive dysfunction therapy, as a causative agent. If this patient had multiple prescriptions, donepezil was probably not considered a major cause of hypokalemia. Donepezil is a cholinesterase inhibitor, mainly prescribed for Alzheimer disease. By inhibiting acetylcholinesterase, donepezil improves behavioral and cognitive symptoms, including confusion, aggression, and psychosis [11,12]. In several studies, donepezil has demonstrated improved cognitive functions in patients with dementia, but it had some adverse effects [13]. The adverse effects were associated with increased cholinergic activity, and the gastrointestinal system was mainly affected. Therefore, nausea, vomiting, and diarrhea were the most common symptoms, as well as insomnia, abnormal dreams, hepatotoxicity, and cardiovascular adverse events [14]. Hypokalemia was one of the rare adverse effects. The mechanism by which donepezil causes hypokalemia remains unclear. In this case, based on TTKG, potassium excretion continued under donepezil therapy. Therefore, we hypothesized the mechanism by which donepezil caused renal potassium wasting. This finding may be due to the action of donepezil on the ion channels in the renal tubule or Henle's loop. Donepezil could potentially inhibit the Na+-K+-2Cl- cotransporter in the thick ascending limb of Henle's loop, suppressing potassium reabsorption. Additionally, it could be postulated that donepezil stimulates the renal outer medullary potassium channel to excrete potassium. As another hypothesis, donepezil could affect the sodium channel epithelium in the principal cell of the collecting tubule. Thereafter, potassium appeared to be secreted into the lumen to maintain electrolyte balance. Although the mechanism by which donepezil induces hypokalemia remains unclear, we anticipated that renal potassium wasting would recover after its withdrawal. Unfortunately, the follow-up study did not achieve demonstrate an improvement in renal potassium wasting. Based on previous reports, we speculated the possible reasons by which recovery from renal potassium wasting may vary from 2 days to 3 months [15,16]. Therefore, the patient may require further time for recovery. Nonetheless, this case was significant as it confirmed the progression of hypokalemia recovery, necessitating a decrease in the daily amount of potassium supplementation after the withdrawal of donepezil. A limitation of this case report is that we have not done additional research regarding how donepezil affects the renal ion channels. Further case reports and research would assist in determining the mechanism by which donepezil causes renal potassium wasting. Although donepezil demonstrates rare serious adverse events, symptoms during early therapy require a differential diagnosis for hypokalemia. Conflicts of interest No potential conflict of interest relevant to this article was reported. Author contributions Conceptualization: all authors; Data curation: DK, THB, HSP; Formal analysis: DK, THB, HEY, BSC, BSK; Investigation: DK, THB, HEY, HSP; Methodology, Project administration: THB; Supervision: THB, BSC, BSK; Validation: THB, SJS, BSC, BSK; Writing-original draft: DK; Writing-review & editing: DK, THB, HEY, HSP, SJS. Fig. 1. Abdominal computed tomography scan. (A) Both adrenal glands (arrows) show no abnormalities. (B) Both kidneys (arrows) show no abnormal findings. Fig. 2. Summary of serum potassium level, potassium supplementation, total intake, and total output per day. IV, intravenous; PO, per oral.
5 mg (milligrams).
DrugDosage
CC BY-NC
33045804
18,430,605
2021-01
What was the outcome of reaction 'Hypokalaemia'?
Development of donepezil-induced hypokalemia following treatment of cognitive impairment. Donepezil is a cholinesterase inhibitor used extensively to treat Alzheimer disease. The increased cholinergic activity is associated with adverse effects, therefore gastrointestinal symptoms, including nausea, vomiting, and diarrhea, are common. Hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients. Although hypokalemia of mild and moderate grade does not present serious signs and symptoms, severe hypokalemia often results in prolonged hospitalization and mortality. Herein, we report a case of hypokalemia developed after the initiation of donepezil therapy for cognitive impairment. Introduction Hypokalemia is a common electrolyte disturbance in clinical practice. The major causes of hypokalemia include gastrointestinal loss and medications such as diuretics [1,2]. Most cases are asymptomatic and mild, but some patients develop severe hypokalemia resulting in arrhythmias and patient death. About 20% of inpatients experience hypokalemia during hospitalization [3], and among these cases, severe hypokalemia was sometimes associated with prolonged hospitalization and increased mortality [4]. Donepezil is the second approved acetylcholinesterase inhibitor for the treatment of mild to moderate Alzheimer disease by the United States Food and Drug Administration (FDA), extensively used worldwide [5]. As donepezil is generally tolerated, most adverse events are gastrointestinal symptoms, including vomiting and diarrhea [6]. However, rare adverse events such as lupus, psychosis, and arrhythmia have been reported in a few patients [7,8]. According to the FDA, hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients [9,10]. Notably, hypokalemia exhibits non-specific symptoms such as general weakness, fatigue, dyspepsia, myalgia, tingling sensation, muscle cramps, and spasms; hence, it can be difficult to diagnose without laboratory investigations. Therefore, clinicians sometimes fail to recognize the signs and symptoms of hypokalemia. Accordingly, a rare case of hypokalemia induced by medication is significant. Herein, we report a case of hypokalemia developed after initiating donepezil as a treatment for cognitive impairment. Case The study was approved by the Institutional Review Board of the Catholic University of Korea (IRB No: PC20ZASI0046) with waiver of informed consent. An 87-year-old man visited the outpatient clinic owing to poor oral intake. According to his medical history, he had undergone surgery for benign prostatic hyperplasia 6 years ago and had discontinued the medication for benign prostatic hyperplasia 4 months before hospitalization. Recently, he was diagnosed with cognitive dysfunction based on a mini-mental status examination score of 22, and a global deterioration scale of 3. And was started on appropriate medication. At the time of admission, he was prescribed choline alfoscerate 400 mg twice daily and donepezil 5 mg for the control of Alzheimer disease. A review of systems did not reveal abnormal findings. His vital signs were as follows: blood pressure (BP), 180/70 mmHg; pulse rate, 72 beats/min; respiratory rate, 20 breaths/min; body temperate, 36.7℃. To determine the cause of poor oral intake, routine investigations were performed, including complete blood count, blood chemistry, urine analysis, electrocardiogram, and chest X-ray. The presence of subtle pneumonia on the chest X-ray and hypokalemia (serum potassium 2.6 mmol/L) was detected. Initial serum inflammatory markers showed the following results: plasma leukocyte count, 7,500/mm3; serum c-reactive protein, 0.74 mg/dL (range, 0.01–0.5 mg/dL). Except for potassium, blood chemistry revealed the following: serum albumin, 4.0 g/dL (range, 3.5–5.2 g/dL); blood urea nitrogen, 10.9 mg/dL; serum creatinine, 0.72 mg/dL (range, 0.61–1.20 mg/dL); serum sodium, 140 mmol/L; serum chloride, 91 mmol/L; serum magnesium, 2.4 mg/dL; serum osmolality, 290 mOsm/kg. Urinalysis and urine sediment examination presented the following: urine pH, 7.5; urine specific gravity, 1.011; urine white blood cells, 0–2/high power field; urine red blood cells, 0–2/high power field. Blood gas analysis revealed compensated metabolic alkalosis as follows: arterial blood pH, 7.52; arterial blood pCO2, 46.6 mmHg; arterial blood pO2, 73.7 mmHg; arterial blood HCO3¯, 36.8 mmol/L. Chest X-ray and inflammatory markers suggested that the pneumonia was not severe. Therefore, hypokalemia could be the cause of underlying poor oral intake. The patient was admitted to treat pneumonia and hypokalemia. For pneumonia treatment, he received ceftriaxone and clarithromycin as antibiotic agents. To determine the cause of hypokalemia, further investigations were performed. No clues indicating hypokalemia were detected in the medical history and review of systems. Next, his renal potassium excretion was measured to determine the potassium/creatinine ratio, fractional excretion of potassium, and transtubular potassium gradient (TTKG), using blood chemistry and spot urine chemistry. The results of spot urine chemistry were as follows: urea nitrogen, 392.8 mg/dL; creatinine, 90.8 mg/dL; total protein, 29.9 mg/dL; sodium, 35 mEq/L; potassium, 29.2 mEq/L; chloride, 45 mEq/L; osmolality, 309 mOsm/kg. Additionally, the following values were determined and suggested renal potassium wasting: urine potassium/creatinine ratio, 32.2 mEq/g; fractional excretion of potassium, 8.9%; TTKG, 10.5. Those findings suggested renal potassium wasting. Next, computed tomography demonstrated no abnormalities in the bilateral adrenal glands and both kidneys (Fig. 1). A hormone study was conducted to evaluate the cause of high BP. The thyroid function test was in the normal range. The adrenocorticotropic hormone (ACTH) level was 36.02 pg/mL (range, 10.0–60.0 pg/mL) and the cortisol level was 10.51 µg/dL (range, 9.41–26.06 µg/dL), which were within the normal range, revealing no mineral corticoid excess. Plasma renin activity was determined as 0.19 ng/mL/hr (range, 0.3–2.9 ng/mL/hr), aldosterone concentration was 14.93 pg/mL (range, 29.9–158.8 pg/mL), and the aldosterone/renin ratio was 7.86, indicating hyporeninemic hypoaldosteronism. However, unlike Liddle syndrome, the serum sodium level was normal at 140 mmol/L. The serum magnesium level was normal at 2.4 mg/dL, which was far from that observed in Gitelman syndrome. Hence, no obvious cause of hypokalemia was determined. During investigations analyzing the possible cause of hypokalemia, he underwent potassium supplementation using intravenous and oral formulations. Until the fourth day of hospitalization, his serum potassium level demonstrated difficulty increasing beyond 3.0 mmol/L despite sufficient potassium administration (Fig. 2). As the hypokalemia was poorly corrected, clinicians suspected other causes of hypokalemia. A thorough review of the therapeutic agents prescribed for cognitive dysfunction was conducted. On examining possible adverse effects, we identified evidence suggesting that donepezil causes hypokalemia as a rare adverse event [9,10]. Therefore, donepezil was immediately discontinued. Thereafter, his potassium demand, supplemented intravenously, gradually decreased and was modified to oral potassium agents. On the third day after donepezil cessation, his serum potassium level recovered to 3.5 mmol/L under oral potassium supplementation of 32 mEq/day (Fig. 2). During hypokalemia evaluation and correction, pneumonia was properly controlled. Finally, he was able to maintain serum potassium of 3.3 mmol/L under oral potassium supplementation of 32 mEq/day and was discharged on the ninth day of hospitalization. At the time of discharge, the results of blood chemistry and spot urine chemistry were as follows: serum creatinine, 0.73 mg/dL; serum potassium, 3.5 mmol/L; serum magnesium, 2.3 mg/dL; serum osmolality, 295 mOsm/kg; urine creatinine, 120.6 mg/dL; potassium, 46.7 mEq/L; osmolality, 330 mOsm/kg. Additionally, the following values were determined: urine potassium/creatinine ratio, 38.7 mEq/g; fractional excretion of potassium, 8.1%; TTKG, 11.9. Conversely, in the past, he had presented a systolic BP of less than 130 mmHg and diastolic BP of 80 mmHg during several visits to the outpatient clinic, with no history of medication-related hypertension. However, for 24 hours after hospitalization, BP was continuously confirmed as 160/90 mmHg or more, including a maximum of 185/105 mmHg, and amlodipine 5 mg was initiated. As BP measurements were above 140/90 mmHg on average after the addition of amlodipine 5 mg, the patient was additionally prescribed olmesartan 20 mg on the fifth day of hospitalization. Finally, in the outpatient clinic follow-ups, serum potassium was measured as 4.5 mmol/L, and oral potassium agents were withheld. Discussion This case report presents the rare development of hypokalemia with donepezil, an agent used to treat cognitive dysfunction. The most common cause of hypokalemia is gastrointestinal losses, followed by medications such as diuretics [1,2]. Additionally, various conditions lead to hypokalemia. For example, the following factors result in the development of renal potassium wasting: malignant hypertension, renal artery stenosis, renin-secreting tumors that can increase renin, adrenal hyperplasia, Cushing syndrome, medication including diuretics, magnesium deficiency, Gitelman syndrome, and chronic metabolic acidosis [3]. However, it is difficult to implicate donepezil, used for cognitive dysfunction therapy, as a causative agent. If this patient had multiple prescriptions, donepezil was probably not considered a major cause of hypokalemia. Donepezil is a cholinesterase inhibitor, mainly prescribed for Alzheimer disease. By inhibiting acetylcholinesterase, donepezil improves behavioral and cognitive symptoms, including confusion, aggression, and psychosis [11,12]. In several studies, donepezil has demonstrated improved cognitive functions in patients with dementia, but it had some adverse effects [13]. The adverse effects were associated with increased cholinergic activity, and the gastrointestinal system was mainly affected. Therefore, nausea, vomiting, and diarrhea were the most common symptoms, as well as insomnia, abnormal dreams, hepatotoxicity, and cardiovascular adverse events [14]. Hypokalemia was one of the rare adverse effects. The mechanism by which donepezil causes hypokalemia remains unclear. In this case, based on TTKG, potassium excretion continued under donepezil therapy. Therefore, we hypothesized the mechanism by which donepezil caused renal potassium wasting. This finding may be due to the action of donepezil on the ion channels in the renal tubule or Henle's loop. Donepezil could potentially inhibit the Na+-K+-2Cl- cotransporter in the thick ascending limb of Henle's loop, suppressing potassium reabsorption. Additionally, it could be postulated that donepezil stimulates the renal outer medullary potassium channel to excrete potassium. As another hypothesis, donepezil could affect the sodium channel epithelium in the principal cell of the collecting tubule. Thereafter, potassium appeared to be secreted into the lumen to maintain electrolyte balance. Although the mechanism by which donepezil induces hypokalemia remains unclear, we anticipated that renal potassium wasting would recover after its withdrawal. Unfortunately, the follow-up study did not achieve demonstrate an improvement in renal potassium wasting. Based on previous reports, we speculated the possible reasons by which recovery from renal potassium wasting may vary from 2 days to 3 months [15,16]. Therefore, the patient may require further time for recovery. Nonetheless, this case was significant as it confirmed the progression of hypokalemia recovery, necessitating a decrease in the daily amount of potassium supplementation after the withdrawal of donepezil. A limitation of this case report is that we have not done additional research regarding how donepezil affects the renal ion channels. Further case reports and research would assist in determining the mechanism by which donepezil causes renal potassium wasting. Although donepezil demonstrates rare serious adverse events, symptoms during early therapy require a differential diagnosis for hypokalemia. Conflicts of interest No potential conflict of interest relevant to this article was reported. Author contributions Conceptualization: all authors; Data curation: DK, THB, HSP; Formal analysis: DK, THB, HEY, BSC, BSK; Investigation: DK, THB, HEY, HSP; Methodology, Project administration: THB; Supervision: THB, BSC, BSK; Validation: THB, SJS, BSC, BSK; Writing-original draft: DK; Writing-review & editing: DK, THB, HEY, HSP, SJS. Fig. 1. Abdominal computed tomography scan. (A) Both adrenal glands (arrows) show no abnormalities. (B) Both kidneys (arrows) show no abnormal findings. Fig. 2. Summary of serum potassium level, potassium supplementation, total intake, and total output per day. IV, intravenous; PO, per oral.
Recovered
ReactionOutcome
CC BY-NC
33045804
18,430,605
2021-01
What was the outcome of reaction 'Pneumonia'?
Development of donepezil-induced hypokalemia following treatment of cognitive impairment. Donepezil is a cholinesterase inhibitor used extensively to treat Alzheimer disease. The increased cholinergic activity is associated with adverse effects, therefore gastrointestinal symptoms, including nausea, vomiting, and diarrhea, are common. Hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients. Although hypokalemia of mild and moderate grade does not present serious signs and symptoms, severe hypokalemia often results in prolonged hospitalization and mortality. Herein, we report a case of hypokalemia developed after the initiation of donepezil therapy for cognitive impairment. Introduction Hypokalemia is a common electrolyte disturbance in clinical practice. The major causes of hypokalemia include gastrointestinal loss and medications such as diuretics [1,2]. Most cases are asymptomatic and mild, but some patients develop severe hypokalemia resulting in arrhythmias and patient death. About 20% of inpatients experience hypokalemia during hospitalization [3], and among these cases, severe hypokalemia was sometimes associated with prolonged hospitalization and increased mortality [4]. Donepezil is the second approved acetylcholinesterase inhibitor for the treatment of mild to moderate Alzheimer disease by the United States Food and Drug Administration (FDA), extensively used worldwide [5]. As donepezil is generally tolerated, most adverse events are gastrointestinal symptoms, including vomiting and diarrhea [6]. However, rare adverse events such as lupus, psychosis, and arrhythmia have been reported in a few patients [7,8]. According to the FDA, hypokalemia is a rare adverse event that occurs in less than 1% of donepezil-treated patients [9,10]. Notably, hypokalemia exhibits non-specific symptoms such as general weakness, fatigue, dyspepsia, myalgia, tingling sensation, muscle cramps, and spasms; hence, it can be difficult to diagnose without laboratory investigations. Therefore, clinicians sometimes fail to recognize the signs and symptoms of hypokalemia. Accordingly, a rare case of hypokalemia induced by medication is significant. Herein, we report a case of hypokalemia developed after initiating donepezil as a treatment for cognitive impairment. Case The study was approved by the Institutional Review Board of the Catholic University of Korea (IRB No: PC20ZASI0046) with waiver of informed consent. An 87-year-old man visited the outpatient clinic owing to poor oral intake. According to his medical history, he had undergone surgery for benign prostatic hyperplasia 6 years ago and had discontinued the medication for benign prostatic hyperplasia 4 months before hospitalization. Recently, he was diagnosed with cognitive dysfunction based on a mini-mental status examination score of 22, and a global deterioration scale of 3. And was started on appropriate medication. At the time of admission, he was prescribed choline alfoscerate 400 mg twice daily and donepezil 5 mg for the control of Alzheimer disease. A review of systems did not reveal abnormal findings. His vital signs were as follows: blood pressure (BP), 180/70 mmHg; pulse rate, 72 beats/min; respiratory rate, 20 breaths/min; body temperate, 36.7℃. To determine the cause of poor oral intake, routine investigations were performed, including complete blood count, blood chemistry, urine analysis, electrocardiogram, and chest X-ray. The presence of subtle pneumonia on the chest X-ray and hypokalemia (serum potassium 2.6 mmol/L) was detected. Initial serum inflammatory markers showed the following results: plasma leukocyte count, 7,500/mm3; serum c-reactive protein, 0.74 mg/dL (range, 0.01–0.5 mg/dL). Except for potassium, blood chemistry revealed the following: serum albumin, 4.0 g/dL (range, 3.5–5.2 g/dL); blood urea nitrogen, 10.9 mg/dL; serum creatinine, 0.72 mg/dL (range, 0.61–1.20 mg/dL); serum sodium, 140 mmol/L; serum chloride, 91 mmol/L; serum magnesium, 2.4 mg/dL; serum osmolality, 290 mOsm/kg. Urinalysis and urine sediment examination presented the following: urine pH, 7.5; urine specific gravity, 1.011; urine white blood cells, 0–2/high power field; urine red blood cells, 0–2/high power field. Blood gas analysis revealed compensated metabolic alkalosis as follows: arterial blood pH, 7.52; arterial blood pCO2, 46.6 mmHg; arterial blood pO2, 73.7 mmHg; arterial blood HCO3¯, 36.8 mmol/L. Chest X-ray and inflammatory markers suggested that the pneumonia was not severe. Therefore, hypokalemia could be the cause of underlying poor oral intake. The patient was admitted to treat pneumonia and hypokalemia. For pneumonia treatment, he received ceftriaxone and clarithromycin as antibiotic agents. To determine the cause of hypokalemia, further investigations were performed. No clues indicating hypokalemia were detected in the medical history and review of systems. Next, his renal potassium excretion was measured to determine the potassium/creatinine ratio, fractional excretion of potassium, and transtubular potassium gradient (TTKG), using blood chemistry and spot urine chemistry. The results of spot urine chemistry were as follows: urea nitrogen, 392.8 mg/dL; creatinine, 90.8 mg/dL; total protein, 29.9 mg/dL; sodium, 35 mEq/L; potassium, 29.2 mEq/L; chloride, 45 mEq/L; osmolality, 309 mOsm/kg. Additionally, the following values were determined and suggested renal potassium wasting: urine potassium/creatinine ratio, 32.2 mEq/g; fractional excretion of potassium, 8.9%; TTKG, 10.5. Those findings suggested renal potassium wasting. Next, computed tomography demonstrated no abnormalities in the bilateral adrenal glands and both kidneys (Fig. 1). A hormone study was conducted to evaluate the cause of high BP. The thyroid function test was in the normal range. The adrenocorticotropic hormone (ACTH) level was 36.02 pg/mL (range, 10.0–60.0 pg/mL) and the cortisol level was 10.51 µg/dL (range, 9.41–26.06 µg/dL), which were within the normal range, revealing no mineral corticoid excess. Plasma renin activity was determined as 0.19 ng/mL/hr (range, 0.3–2.9 ng/mL/hr), aldosterone concentration was 14.93 pg/mL (range, 29.9–158.8 pg/mL), and the aldosterone/renin ratio was 7.86, indicating hyporeninemic hypoaldosteronism. However, unlike Liddle syndrome, the serum sodium level was normal at 140 mmol/L. The serum magnesium level was normal at 2.4 mg/dL, which was far from that observed in Gitelman syndrome. Hence, no obvious cause of hypokalemia was determined. During investigations analyzing the possible cause of hypokalemia, he underwent potassium supplementation using intravenous and oral formulations. Until the fourth day of hospitalization, his serum potassium level demonstrated difficulty increasing beyond 3.0 mmol/L despite sufficient potassium administration (Fig. 2). As the hypokalemia was poorly corrected, clinicians suspected other causes of hypokalemia. A thorough review of the therapeutic agents prescribed for cognitive dysfunction was conducted. On examining possible adverse effects, we identified evidence suggesting that donepezil causes hypokalemia as a rare adverse event [9,10]. Therefore, donepezil was immediately discontinued. Thereafter, his potassium demand, supplemented intravenously, gradually decreased and was modified to oral potassium agents. On the third day after donepezil cessation, his serum potassium level recovered to 3.5 mmol/L under oral potassium supplementation of 32 mEq/day (Fig. 2). During hypokalemia evaluation and correction, pneumonia was properly controlled. Finally, he was able to maintain serum potassium of 3.3 mmol/L under oral potassium supplementation of 32 mEq/day and was discharged on the ninth day of hospitalization. At the time of discharge, the results of blood chemistry and spot urine chemistry were as follows: serum creatinine, 0.73 mg/dL; serum potassium, 3.5 mmol/L; serum magnesium, 2.3 mg/dL; serum osmolality, 295 mOsm/kg; urine creatinine, 120.6 mg/dL; potassium, 46.7 mEq/L; osmolality, 330 mOsm/kg. Additionally, the following values were determined: urine potassium/creatinine ratio, 38.7 mEq/g; fractional excretion of potassium, 8.1%; TTKG, 11.9. Conversely, in the past, he had presented a systolic BP of less than 130 mmHg and diastolic BP of 80 mmHg during several visits to the outpatient clinic, with no history of medication-related hypertension. However, for 24 hours after hospitalization, BP was continuously confirmed as 160/90 mmHg or more, including a maximum of 185/105 mmHg, and amlodipine 5 mg was initiated. As BP measurements were above 140/90 mmHg on average after the addition of amlodipine 5 mg, the patient was additionally prescribed olmesartan 20 mg on the fifth day of hospitalization. Finally, in the outpatient clinic follow-ups, serum potassium was measured as 4.5 mmol/L, and oral potassium agents were withheld. Discussion This case report presents the rare development of hypokalemia with donepezil, an agent used to treat cognitive dysfunction. The most common cause of hypokalemia is gastrointestinal losses, followed by medications such as diuretics [1,2]. Additionally, various conditions lead to hypokalemia. For example, the following factors result in the development of renal potassium wasting: malignant hypertension, renal artery stenosis, renin-secreting tumors that can increase renin, adrenal hyperplasia, Cushing syndrome, medication including diuretics, magnesium deficiency, Gitelman syndrome, and chronic metabolic acidosis [3]. However, it is difficult to implicate donepezil, used for cognitive dysfunction therapy, as a causative agent. If this patient had multiple prescriptions, donepezil was probably not considered a major cause of hypokalemia. Donepezil is a cholinesterase inhibitor, mainly prescribed for Alzheimer disease. By inhibiting acetylcholinesterase, donepezil improves behavioral and cognitive symptoms, including confusion, aggression, and psychosis [11,12]. In several studies, donepezil has demonstrated improved cognitive functions in patients with dementia, but it had some adverse effects [13]. The adverse effects were associated with increased cholinergic activity, and the gastrointestinal system was mainly affected. Therefore, nausea, vomiting, and diarrhea were the most common symptoms, as well as insomnia, abnormal dreams, hepatotoxicity, and cardiovascular adverse events [14]. Hypokalemia was one of the rare adverse effects. The mechanism by which donepezil causes hypokalemia remains unclear. In this case, based on TTKG, potassium excretion continued under donepezil therapy. Therefore, we hypothesized the mechanism by which donepezil caused renal potassium wasting. This finding may be due to the action of donepezil on the ion channels in the renal tubule or Henle's loop. Donepezil could potentially inhibit the Na+-K+-2Cl- cotransporter in the thick ascending limb of Henle's loop, suppressing potassium reabsorption. Additionally, it could be postulated that donepezil stimulates the renal outer medullary potassium channel to excrete potassium. As another hypothesis, donepezil could affect the sodium channel epithelium in the principal cell of the collecting tubule. Thereafter, potassium appeared to be secreted into the lumen to maintain electrolyte balance. Although the mechanism by which donepezil induces hypokalemia remains unclear, we anticipated that renal potassium wasting would recover after its withdrawal. Unfortunately, the follow-up study did not achieve demonstrate an improvement in renal potassium wasting. Based on previous reports, we speculated the possible reasons by which recovery from renal potassium wasting may vary from 2 days to 3 months [15,16]. Therefore, the patient may require further time for recovery. Nonetheless, this case was significant as it confirmed the progression of hypokalemia recovery, necessitating a decrease in the daily amount of potassium supplementation after the withdrawal of donepezil. A limitation of this case report is that we have not done additional research regarding how donepezil affects the renal ion channels. Further case reports and research would assist in determining the mechanism by which donepezil causes renal potassium wasting. Although donepezil demonstrates rare serious adverse events, symptoms during early therapy require a differential diagnosis for hypokalemia. Conflicts of interest No potential conflict of interest relevant to this article was reported. Author contributions Conceptualization: all authors; Data curation: DK, THB, HSP; Formal analysis: DK, THB, HEY, BSC, BSK; Investigation: DK, THB, HEY, HSP; Methodology, Project administration: THB; Supervision: THB, BSC, BSK; Validation: THB, SJS, BSC, BSK; Writing-original draft: DK; Writing-review & editing: DK, THB, HEY, HSP, SJS. Fig. 1. Abdominal computed tomography scan. (A) Both adrenal glands (arrows) show no abnormalities. (B) Both kidneys (arrows) show no abnormal findings. Fig. 2. Summary of serum potassium level, potassium supplementation, total intake, and total output per day. IV, intravenous; PO, per oral.
Recovering
ReactionOutcome
CC BY-NC
33045804
18,742,345
2021-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'No adverse event'.
Retrospective Review of Pharmacogenetic Testing at an Academic Children's Hospital. There is limited evidence to support pharmacogenetic (PGx) testing in children. We conducted a retrospective review of PGx testing among 452 patients at an academic children's hospital to determine the potential utility of PGx in diseases of childhood and to identify targets for future pediatric pharmacogenetic research. An actionable gene-drug pair associated with the 28 genes tested (Clinical Pharmacogenetics Implementation Consortium (CPIC) level A or B, Pharmacogenomics Knowledge Base (PharmGKB) level 1A or B, or US Food and Drug Administration (FDA) recommendation and a PharmGKB level) was present in 98.7% of patients. We identified 203 actionable gene-drug-diagnosis groups based on the indications for each actionable drug listed in Lexicomp. Among patients with an actionable gene-drug-diagnosis group, 49.3% had a diagnosis where the drug was a therapeutic option and PGx could be used to guide treatment selection. Among patients with an associated diagnosis, 30.9% had a prescription for the actionable drug allowing PGx guided dosing. Three genes (CYP2C19, CYP2D6, and CYP3A5) accounted for all the gene-drug-diagnosis groups with matching diagnoses and prescriptions. The most common gene-drug-diagnosis groups with matching diagnoses and prescriptions were CYP2C19-citalopram-escitalopram-depression 3.3% of patients tested; CYP2C19-dexlansoprazole-gastritis-esophagitis 3.1%; CYP2C19-omeprazole-gastritis-esophagitis 2.4%; CYP2D6-atomoxetine-attention deficit hyperactivity disorder 2.2%; and CYP2C19-citalopram-escitalopram-obsessive-compulsive disorder 1.5%. PGx could be used to guide selection of current treatment options or medication dosing in almost half (48.7%) of pediatric patients tested. Mood disorders and gastritis/esophagitis are promising targets for future study of PGx testing because of the high prevalence of these diagnoses and associated actionable gene-drug pairs in the pediatric population. Study Highlights WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? There is limited evidence supporting the clinical utility of pharmacogenetic (PGx) testing in children. WHAT QUESTION DID THIS STUDY ADDRESS? Evaluate the potential clinical utility of PGx testing performed on children to assist with treatment selection and dose adjustment and to identify targets for future research. WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE? This study improves our understanding of the potential use of PGx testing in treatment selection and dose adjustment while treating children. Almost half of our patients (47.8%) had a clinical diagnosis where their results could influence treatment and 15.0% were prescribed a medication where their test results could be used to adjust dosing. Three genes (CYP2C19, CYP2D6, and CYP3A5) accounted for all the actionable gene‐drug pairs with matching diagnoses and prescriptions. Mood‐disorders‐selective serotonin reuptake inhibitor and gastroesophagitis‐proton pump inhibitors were the most commonly affected diagnosis‐drug combinations identified. HOW MIGHT THIS CHANGE CLINICAL PHARMACOLOGY OR TRANSLATIONAL SCIENCE? Results from this study identify areas for future PGx research in children and may guide the development of tailored clinical decision support tools to better serve their needs. Pharmacogenetic (PGx) testing, specifically the inquiry into genetic variants in pharmacokinetic or pharmacodynamic pathways involved in medication metabolism or response, is commercially available and being promoted to improve patient outcomes. 1 The level of evidence supporting the clinical utility of testing for variants in individual genes differs, and translation of the test results into clinical practice is complicated. Several organizations, including the Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Royal Dutch Association for the Advancement of Pharmacy – Pharmacogenetics Working Group (DPWG), have created detailed, evidence‐based, gene‐drug clinical practice guidelines that can assist providers in applying the results of PGx testing to the management of medications used to treat mood disorders, cardiovascular disease, cancer, or other diseases among adults. 2 , 3 Previous research in adult populations has demonstrated the clinical utility 4 , 5 , 6 , 7 , 8 and cost‐effectiveness 9 , 10 for both reactive PGx testing to guide current treatment and pre‐emptive testing to guide future treatment decisions. However, the evidence base for PGx testing in pediatric patients and in treatment of diseases prevalent during childhood is less robust. Despite the lack of current evidence in favor of PGx implementation for children, enthusiasm for the potential of PGx remains high among pediatric clinicians. A recent survey of pediatric providers in the United States and Japan found that > 80% believe PGx will improve the safety and efficacy of pediatric drug therapy. Findings from this survey endorsed the interest in education to equip pediatric clinicians with the skills to implement PGx. 11 However, the current guidelines for interpreting and implementing PGx are based largely on adult data, and it is unclear how well they apply to the care of children and adolescents. Thus, PGx testing in pediatrics is most often reactive, and it is not clear which pediatric patients may benefit most from reactive testing with regard to their current clinical care or pre‐emptive testing to guide future drug therapy. A previous study of medication use at a children’s hospital, identified 10 commonly prescribed medications with evidence‐based guidelines for dosing adjustment based on PGx testing results (ondansetron, oxycodone, codeine, omeprazole, lansoprazole, sertraline, amitriptyline, citalopram, escitalopram, and risperidone). Only three of these drugs (codeine, omeprazole, and lansoprazole) had adequate pediatric data in support of age‐specific recommendations. 12 The goal of this study was to describe the proportion of pediatric age patients who may benefit from pre‐emptive PGx testing and identify high‐impact areas for future research by conducting a retrospective review of the records of patients who had PGx testing performed at Children’s Mercy Hospital. We identified (1) actionable gene‐drug pairs associated with the genes evaluated at our hospital, (2) clinical diagnostic groups associated with these gene‐drug pairs creating gene‐drug‐diagnosis groups, (3) the proportion of patients who can use PGx testing results to assist with treatment selection for current diagnoses (potentially actionable gene‐drug‐diagnosis group and a matching clinical diagnosis, (4) the proportion of patients who can use PGx testing results to guide medication dosing (potentially actionable gene‐drug‐diagnosis group with a matching clinical diagnosis and drug prescription), and (5) the pediatric diagnosis groups with the highest frequency of patients with actionable test results with a matching clinical diagnosis and drug prescription. METHODS This is a retrospective review of PGx testing of pediatric patients conducted between 2017 and 2019 at Children’s Mercy Hospital, an academic children’s hospital in the midwestern United States. Testing could be obtained by community providers through referral to the Genomic and Ontogeny‐Linked Dose Individualization and cLinical Optimization for KidS (GOLDILOKs) Clinic or by Children’s Mercy providers ordering the test directly. The hospital used a commercially available, 20‐gene PGx panel (OneOme, Minneapolis, MN) for all tests. The specific genes assessed on this panel varied over time, as particular genes were removed and replaced with other genes, so the results from 28 different genes were assessed. When the results of this test were returned to Children’s Mercy from the outside laboratory, one of the providers trained in PGx reviewed the results. Then, that provider sent the family and the provider who requested the test an individualized explanation of the results in addition to relevant specific medication guidance. The testing was performed at the discretion of the ordering provider. There was no clinical decision support built into the electronic health record (EHR) based on the results of the PGx testing, and testing was not performed as part of a structured pre‐emptive testing program. In addition to PGx test results, we also collected the age, sex, and race/ethnicity for each patient. To determine both the current and possible future clinical utility of PGx testing in the patients tested, we reviewed the test results from each patient to identify potentially actionable genetic variants with evidence‐based guidance for drug therapy (gene‐drug pairs). We also identified clinical diagnoses for which the drug guidance can be put into action (gene‐drug‐diagnosis groups). We defined potentially actionable gene‐drug pairs at the allele level as the following: (i) CPIC level A or B guidance, (ii) Pharmacogenomics Knowledge Base (PharmGKB) level 1A or 1B guidance, or (iii) gene‐drug pairs with a US Food and Drug Administration (FDA)‐approved drug label of “actionable pgx,” “genetic testing recommended,” or “genetic testing required” and a PharmGKB PGx level. We only searched for guidelines or recommendations associated with the alleles that were present in our sample population. For each actionable gene‐drug pair, we identified all FDA‐approved and off‐label indications for the drug using the Lexicomp medication database if the PGx guidance did not limit treatment recommendations to a specific disease process. 13 Both adult and pediatric indications for medications were included in our study. These indications were subsequently mapped to a specific diagnosis or group of diagnoses and their associated International Classification of Disease‐10th edition (ICD‐10) codes. We combined the clinical indications and associated diagnoses for each drug with our list of potentially actionable gene‐drug pairs to create a list of potentially actionable gene‐drug‐diagnosis groups. We included both off‐label and FDA‐approved indications because off‐label medication use is a frequent occurrence in pediatric care. 14 We did not include conditions that we were unable to map to a limited set of ICD‐10 codes that would make an effective target for pre‐emptive PGx testing, such as acute pain or severe nausea. 15 We also did not include drugs where the guidelines available at the time of the study did not provide any dosing recommendations based on PGx test results (e.g., risperidone). We initially included SLC6A4:citalopram/escitalopram in our study as this had a CPIC level of B/C and dosage guidance in PharmGKB. However, the evidence supporting this dosage guidance was contradictory at the time of our initial review of the CPIC and PharmGKB guidance and has become more unclear over time. Therefore, we elected to remove this gene from our results. For patients with a potentially actionable gene‐drug‐diagnosis group, we reviewed our hospital EHR database containing visit data and ICD‐10 codes to determine if a patient had a current (within 2 years before or after PGx testing) clinical diagnosis with a condition included in that gene‐drug‐diagnosis group. For patients with a matching clinical diagnosis, we reviewed the EHR database to determine if the patient had a prescription documented in the EHR for the drug included in that gene‐drug‐diagnosis group. See Figure 1 for an overview of our analytic strategy. Figure 1 Interpretation of pharmacogenetic test results. CPIC, Clinical Pharmacogenetics Implementation Consortium; FDA, US Food and Drug Administration; PharmGKB, Pharmacogenomics Knowledge Base. Exclusion of medications to treat potential future acute pain or severe nausea will likely underestimate the potential benefits of PGx in general. We attempted, however, to estimate the number of patients per year seen at Children’s Mercy Kansas City who may benefit from PGx testing prior to receiving a prescription for ondansetron, codeine, or oxycodone. First, we determined the number of unique patients prescribed these medications at Children’s Mercy, Kansas City, between April 1, 2018, and March 31, 2019. Then, this number was multiplied by the proportion of patients in our sample with actionable PGx results associated with these drugs. We did not review individual patient notes and attempt to infer the provider’s intent when ordering the PGx testing or how the results were incorporated into the patient’s medical care as this was outside the scope of our study. This study was approved by the Children’s Mercy Kansas City Institutional Review Board. RESULTS PGx test results were available for 452 patients. The average age of our patients was 11.9 ± 4.3 years, 51.8% were boys, and 79.1% were white, 9.9% of African descent, 2.6% Hispanic/Latino, 1.8% Asian, 0.7% American Indian, and 0.7% Hawaiian or Pacific Islander. Ordering services included individualized therapeutics/clinical pharmacology, behavioral health, and adolescent medicine (Table 1 ). Table 1 Study demographics Demographic group Mean ± SD Age, years 11.9 ± 4.3 Sex % of total Female 48.2% Male 51.8% Race/ethnicity White 79.1% Black/African American 9.9% Hispanic/Latino 2.6% Asian 1.8% Native American 0.7% Pacific Islander 0.7% Unknown 5.3% John Wiley & Sons, LtdGene‐drug pairs We identified one or more guidelines or recommendations for drug management associated with 16 of the 28 genes tested on the commercial PGx panel. These guidelines describe a total of 78 potentially actionable gene‐drug pairs associated with the alleles identified by the PGx test. In our sample, 446 of 452 patients (98.7%) tested had at least one potentially actionable gene‐drug pair (Table 2 ). The most common gene‐drug pairs identified were VKORC1‐warfarin with 266 actionable results out of 452 tests (58.8%), IFNL4‐peginterferon alfa‐2b 170 of 292 tests (58.2%), CYP4F2‐warfarin 83 of 187 tests (44.4%), CYP2C19‐dexlansoprazole 148 of 452 tests (32.7%), and CYP2C19‐clopidogrel 148 of 452 tests (32.7%). Table 2 Frequency of actionable PGx test results Gene Percentage of patients tested with an actionable gene‐drug pair Percentage of patients tested where PGx results are useful for treatment of current diseasea Percentage of patients tested where PGx results are useful for dosing of currently prescribed drugsb CYP2C19 62.3 32.9 11.0 CYP2D6 19.6 17.0 4.9 CYP3A5 22.9 0.7 0.2 HLA‐A 5.2 2.6 0.0 CYP2C9 24.9 1.3 0.0 SLCO1B1 27.1 1.1 0.0 TPMT 9.5 0.7 0.0 VKORC1 58.8 0.4 0.0 UGT1A1 11.0 0.2 0.0 NUDT15 2.2 0.2 0.0 IFNL4 58.9 0.0 0.0 CYP4F2 44.0 0.0 0.0 HLA‐B 9.5 0.0 0.0 F5 5.1 0.0 0.0 CYP2C rs12777823 3.4 0.0 0.0 DPYD 2.0 0.0 0.0 HLA‐A alleles assessed: *31:01; HLA‐B alleles assessed: *15:02, *57:01 and *58:01. COMT, CYP1A2, CYP2B6, CYP3A4, DRD2, F2, GRIK4, HTR2A, HTR2C, IL28B, OPRM1, and SLC6A4 genotypes were available on some patients as well, but no evidence‐based guidelines for the interpretation of these results were identified that met our inclusion criteria. PGx, pharmacogenetic. a Patient has ≥ 1 actionable gene‐drug‐diagnosis group and a matching clinical diagnosis within 2 years of testing. b Patient has ≥ 1 actionable gene‐drug‐diagnosis group, a matching clinical diagnosis, and a matching drug prescription within 2 years of testing. John Wiley & Sons, LtdGene‐drug‐diagnosis groups Combining the potentially actionable drug‐gene pairs with the associated FDA‐approved and off‐label indications for the drugs produced 203 gene‐drug‐diagnosis groups (e.g., CYP2C19‐citalopram/escitalopram‐depression). See Table S1 for details of the gene‐drug‐diagnosis groups identified and related ICD‐10 codes. In our sample, the median number of potentially actionable gene‐drug‐diagnosis groups per patient was 20, with a range of 0–109. Matching clinical diagnoses among patients with a potentially actionable gene‐drug‐diagnosis group The PGx test results were informative for treatment selection for current diagnoses among 220 of the 452 patients (48.7%) tested (actionable gene‐drug‐diagnosis group with a matching diagnosis). Ten genes accounted for the gene‐drug‐diagnosis pairs in these patients (Table 2 ). The most common matching clinical diagnosis among patients with a potentially actionable gene‐drug‐diagnosis group was attention deficit‐hyperactivity disorder (ADHD). Of the 193 patients with a gene‐drug‐diagnosis group that included ADHD, 121 (62.7%) had a matching clinical diagnosis of ADHD. Other common conditions with actionable gene‐drug‐diagnosis groups and matching clinical diagnoses included: anxiety disorders 89 of 183 (48.6%), depressive disorders 56 of 183 (30.6%), and gastritis/esophagitis/ulcer disease 56 of 263 (21.3%). Exposure to actionable medications among patients with a potentially actionable gene‐drug‐diagnosis group and a matching clinical diagnosis The PGx results could be used to adjust the dosing of at least one currently prescribed drug among 68 of the 452 patients (15.0%) tested. Two or more potentially actionable gene‐drug‐diagnosis groups with a matching clinical diagnosis and drug prescription within 2 years of testing were present in 22 of 452 patients (4.9%) tested. Three genes accounted for all the gene‐drug pairs involved in this group (CYP2C19, CYP2D6, and CYP3A5; Table 3 ). The most common gene‐drug‐diagnosis groups with a matching clinical diagnosis and prescription among the 452 patients tested were CYP2C19‐citalopram, escitalopram‐depression 3.3% (n = 15); CYP2C19‐dexlansoprazole‐gastritis‐esophagitis 3.1% (n = 14); CYP2C19‐omeprazole‐gastritis‐esophagitis 2.4% (n = 11); CYP2D6‐atomoxetine‐ADHD 2.2% (n = 10); and CYP2C19‐citalopram, escitalopram‐obsessive‐compulsive disorder 1.5% (n = 7). Table 3 Actionable gene‐drug‐diagnosis groups with a matching clinical diagnosis and medication prescription (n = 452) Gene Genotype predicted phenotype Drugs Diagnosis Actionable gene‐drug‐diagnosis group (% of all tested) PGx results are useful for treatment of current disease (% of all tested)a PGx results are useful for dosing of currently prescribed drugs (% of all tested)b CYP2C19 UM (*17/*17) UM/RM (*1/*17) PM (*2/*2, *2/*3) Citalopram, escitalopram Depression 125 (27.7) 34 (7.5) 15 (3.3) Obsessive compulsive disorder 125 (27.7) 22 (4.9) 7 (1.5) Anxiety disorders 125 (27.7) 14 (3.1) 3 (0.7) Citalopram Dementia 125 (27.7) 1 (0.2) 1 (0.2) Escitalopram Autism 125 (27.7) 34 (7.5) 1 (0.2) Clomipramine, doxepin, imipramine Anxiety disorders 125 (27.7) 82 (18.1) 3 (0.7) Amitriptyline, doxepin Insomnia 125 (27.7) 29 (6.4) 3 (0.7) Imipramine ADHD 125 (27.7) 76 (16.8) 2 (0.4) Enuresis 125 (27.7) 8 (1.8) 1 (0.2) Amitriptyline Migraine headache 125 (27.7) 3 (0.7) 1 (0.2) Pantoprazole Gastritis, esophagitis, and ulcers 125 (27.7) 23 (5.1) 6 (1.3) IM (*1/*2, *1/*3, *1/*4, *2/*17) PM (*2/*2, *2/*3) Dexlansoprazole Gastritis, esophagitis, and ulcers 148 (32.7) 34 (7.5) 14 (3.1) UM (*17/*17) UM/RM (*1/*17) Omeprazole Gastritis, esophagitis, and ulcers 115 (25.4) 22 (4.9) 11 (2.4) PM (*2/*2, *2/*3) Lansoprazole Gastritis, esophagitis, and ulcers 10 (2.2) 1 (0.2) 1 (0.2) Sertraline, diazepam Anxiety disorders 10 (2.2) 8 (1.8) 3 (0.7) RM (*1/*17) PM (*2/*2, *2/*3) Amitriptyline Tension headache 95 (21.0) 16 (3.5) 1 (0.2) CYP2D6 UM ((*1/*2A)xN, *1/*1x2, *1/*1xN, *1/*35x2, *2AxN/*2AxN, *2AxN/*41) IM ((*4/*10)xN, *3/*17, *10+*36/*10+*36, *3/*41, *3/*9, *4/*9, *4+*68/*9, *4/*10, *4/*29, *4/*41, *4+*4N/*41, *5/*9, *5/*10+*36, *5/*17, *5/*41, *5/*59, *6/*10, *6/*41) PM ((*4/*4)xN, *3/*4, 3/*6, *4/*4, *4/*4+*68, *4/*5, *4/*6, *5/*68) Amitriptyline, venlafaxine Recurrent headache 76 (16.8) 19 (4.2) 6 (1.3) Amitriptyline, clomipramine, desipramine, doxepin, imipramine, nortriptyline, trimipramine, venlafaxine Depression 76 (16.8) 25 (5.5) 5 (1.1) Clomipramine, venlafaxine Obsessive compulsive disorder 76 (16.8) 16 (3.5) 3 (0.7) Desipramine, imipramine, nortriptyline, venlafaxine ADHD 76 (16.8) 47 (10.4) 2 (0.4) Amitriptyline, doxepin Insomnia 76 (16.8) 15 (3.3) 2 (0.4) Imipramine, venlafaxine Autism 76 (16.8) 20 (4.4) 1 (0.2) Clomipramine, doxepin, imipramine, venlafaxine Anxiety disorders 76 (16.8) 6 (1.3) 1 (0.2) NM (*1/*10, *2A/*10+*36x2, *2A/*10) IM (*10/*10, *10/*29, *10/*41, *10+*36/*10+*36, *3/*9, *3/*17, *3/*41, *4/*9, *4+*68/*9, *4/*10, (*4/*10)xN, *4/*29, *4/*41, *4+*4N/*41, *5/*9, *5/*10+*36, *5/*17, *5/*41, *5/*59, *6/*10, *6/*41) PM ((*4/*4)xN, *3/*4, *3/*6, *4/*4, *4/*4+*68, *4/*5, *4/*6, *5/*68) Atomoxetine ADHD 82 (18.1) 50 (11.1) 10 (2.2) IM (*10/*10, *10/*29, *10/*41, *10+*36/*10+*36, *3/*9, *3/*17, *3/*41, *4/*9, *4+*68/*9, *4/*10, (*4/*10)xN, *4/*29, *4/*41, *4+*4N/*41, *5/*9, *5/*10+*36, *5/*17, *5/*41, *5/*59, *6/*10, *6/*41) PM ((*4/*4)xN, *3/*4, *3/*6, *4/*4, *4/*4+*68, *4/*5, *4/*6, *5/*68) Pimozide Tourette disorder 70 (15.5) 11 (2.4) 2 (0.4) IM (*5/*10+*36) PM (*3/*6, *4/*4, (*4/*4)xN, *4/*4+*68, *4/*5, *4/*6, *5/*68) Aripiprazole, brexpiprazole Depression 30 (6.6) 11 (2.4) 1 (0.2) CYP3A5 NM (*1/*1) IM (*1/*3, *1/*6, *1/*7) Tacrolimus Organ transplant 103 (22.8) 1 (0.2) 1 (0.2) The remaining 183 gene‐drug‐diagnosis groups did not have any patients taking a related medication. Only genotypes that were observed among the cases in our study are listed. Other genotypes (not represented) are likely to be observed in other patient populations. Predicted phenotypes are based on genotype as listed on the OneOme report supplemented by information on the Clinical Pharmacogenetics Implementation Consortium (CPIC) and Pharmacogenomics Knowledge Base (PharmGKB) websites. ADHD, attention deficit hyperactivity disorder; IM, intermediate metabolizer; NM, normal metabolizer (previously referred to as EM, extensive metabolizer); PGx, pharmacogenetic; PM, poor metabolizer; RM, rapid metabolizer; UM, ultrarapid metabolize. 36 Genotypes listed in parentheses (e.g., (*4/*4) × N) indicate the presence of a gene duplication or multiplication on one or both of the alleles. CYP star allele nomenclature is according to the Pharmacogene Variation Consortium (pharmvar.org). 37 , 38 a Patient has ≥ 1 actionable gene‐drug‐diagnosis group and a matching clinical diagnosis within 2 years of testing. b Patient has ≥ 1 actionable gene‐drug‐diagnosis group, a matching clinical diagnosis, and a matching drug prescription within 2 years of testing. John Wiley & Sons, LtdPotential benefit of PGx testing in the treatment of acute pain or severe nausea CYP2D6 ultrarapid metabolizers are at risk for treatment failure when given ondansetron to treat severe nausea. We estimate that 1,227 of the 18,492 patients a year (6.6%) prescribed ondansetron at Children’s Mercy are at risk for treatment failure and could benefit from PGx testing to allow selection of an alternative antinausea medication. CYP2D6 ultrarapid metabolizers are also at risk for respiratory depression when treated with oxycodone. We estimate that ~ 689 of the 10,382 patients a year (6.6%) prescribed oxycodone at Children’s Mercy could benefit from PGx testing to allow selection of an alternative analgesic. Likewise, ~ 4 of the 46 patients a year (8.4%) prescribed codeine could benefit from PGx testing. DISCUSSION Providers could use PGx test results to guide treatment for current diseases in over half of the pediatric patients in our study and to adjust medication dosing in 15% of the patients tested. Most matching diagnoses and drug prescriptions were found among pediatric patients diagnosed with mental health conditions or esophagitis‐gastritis. These two diagnostic groups emerged as the main targets for pre‐emptive testing in children prior to selecting a treatment option or prescribing medication to treat these conditions. Results in context with previous literature The frequency of actionable genetic variants in our study was similar to the frequency seen in previous studies of adult patients. 16 , 17 , 18 This suggests that the underlying genetic diversity in our sample was similar to previous samples. Our study identified many of the same medications (omeprazole, lansoprazole, sertraline, amitriptyline, citalopram, and escitalopram) as a previous study of pediatric age patients. 12 The commonly used medications in one of these studies that we did not identify were specifically excluded from our analysis because we could not map the indications for these drugs, pain and nausea, to a specific set of ICD‐10 codes (ondansetron, oxycodone, and codeine) or because the available guidelines did not provide any guidance on dose modification based on the test results (e.g., risperidone). Thus, our findings provide conservative evidence of the potential clinical utility of PGx testing for conditions outside of acute pain and nausea. We estimate that the inclusion of these broad conditions would increase the apparent benefit PGx testing among pediatric patients. However, identifying and providing pre‐emptive screening to all patients who might experience future severe nausea or acute pain would be logistically challenging. On the other hand, providers who anticipate that their particular patient is at high risk for these conditions should consider PGx testing and include the results in clinical decision support tools to help guide future treatment. Another study examined medication use among children and adults to identify patient populations that would benefit from PGx testing. Like our study, this study identified child mental health as one of the clinical areas most likely to benefit from PGx testing. 19 Our investigation expands on this previous study by examining the potential clinical utility of PGx testing in treatment selection and dose adjustment among pediatric patients by identifying the frequency of matching diagnoses and prescriptions in pediatric patients with a potentially actionable gene‐diagnosis‐drug combination. Although our sample was taken from a select group of patients referred for PGx testing, the immediate clinical utility of the results in our sample was similar to those seen in previous studies of adults and exceeded that reported in pediatric studies. In a study of Chinese children, up to 9% of patients received at least one medication associated with a CPIC guideline. 20 In a study comprising 600 adult patients seen in outpatient or perioperative cardiology clinics and another studying 122 patients with cardiac catheterization, 16.1% and 20% of patients, respectively, were identified to have a PGx variant that may affect the metabolism of a currently taken medication. 21 , 22 When exposure inquiry was expanded to medication use in the past 20 years, 80% of English adults had exposure to at least 1 drug with PGx guidance. 23 This compares to the 15% (68/452) of pediatric patients in our study who had a matching prescription within 2 years before or after PGx testing. Taken together, our results augment current knowledge of the clinical utility of PGx testing in various populations and quantifies the potential impact in pediatric patients. Communicating testing results to pediatric age patients and families Almost all the patients in our sample had a potentially actionable genetic variant. However, only 48.7% had a diagnosis where this information could be clinically useful to current care. For example, of the 452 patients tested, 58.8% had variants of VKORC1 and 44.0% had variants of CYP4F2 that influence warfarin dosing requirements. However, only two of these patients had a clinical diagnosis for which warfarin was a therapeutic option, and neither of them had been prescribed warfarin within 2 years of testing. Describing how “actionable” test results can influence current care and might affect future care in certain circumstances contributes to the complexity of returning PGx results to pediatric patients and their families. Other factors contributing to this complexity include emerging evidence for the possible discordance between adults and children of PGx impact in the setting of obesity. 24 While discussing PGx results, providers need to discuss the impact of test results on current treatment decisions and acknowledge uncertainty when evidence is lacking. Providers also need to discuss test results that do not apply to the patient’s care but need to be preserved in case the results become relevant in the future. Providers also need to be mindful that they do not create additional problems for the child while delivering this information. Parental perception that their child is “abnormal” or has a special susceptibility to problems can result in the vulnerable child syndrome. 25 In this syndrome, unwarranted parental anxiety about a real or perceived illness in a child can change parental behavior causing increased health care utilization, increased anxiety in the child, and limit development of autonomy. Interventions to facilitate discussions of test results and assess provider, patient, and parent understanding of the results would be a valuable area of future research. In addition, studies that examine the risk of increased parental perception of child vulnerability associated with abnormal results against the benefit of providing PGx guidance that applies primarily to diseases prevalent among adults would be beneficial. This is especially true in younger children when the PGx guidance refers to a disease the child is unlikely to have for another 40–50 years, such as breast cancer, and treatment options for that disease which will probably be irrelevant at that point. Potential high‐yield medical conditions for pediatric PGx research Our study revealed that the majority of currently actionable PGx test results are restricted to a small number of genes, thus highlighting the high‐impact, priority areas that should be prioritized in future PGx research endeavors with children. Gastritis/esophagitis/ulcer disease, and mental health disorders were the most frequently identified disorders involved in actionable gene‐drug‐diagnosis groups. These conditions are common problems in pediatric populations, with these gastrointestinal conditions affecting 4.4%, mood disorders affecting 4.2%, and attention deficit disorder/ADHD affecting 8.6% of children and adolescents. 26 , 27 There is also some evidence demonstrating the relevance of PGx testing in the treatment of these conditions in children. CYP2C19 is the enzyme responsible for metabolizing many drugs (e.g., proton pump inhibitors (PPIs) like omeprazole, dex/lansoprazole) prescribed for the treatment of gastritis/ulcer disease. Evidence linking CYP2C19 genotype‐predicted phenotype and PPI adverse events in young children (0–3 years) as well as PPI responsiveness suggests the relevance of this PGx relationship in the pediatric population. 28 , 29 A recent simulation study linked CYP2C19 genotype‐predicted phenotype with altered systemic exposure of the selective serotonin reuptake inhibitors, es/citalopram and sertraline, in children and adolescents. Dose modifications based on genotype‐predicted phenotype (i.e., PGx variation) were suggested, but this has yet to be evaluated prospectively. 30 Other studies have found associations among CYP2C19 genotype‐predicted phenotype and tolerability, adverse events, and time to selective serotonin reuptake inhibitor response among children. 31 , 32 Last, CYP2D6 genotype has been associated with response to psychiatric medications in adults but this has not been confirmed in pediatric patients. 33 Prospective trials of PGx in pediatrics are needed to confirm that the genotype‐predicted phenotype relationships seen in adults are applicable in children and determine the impact of PGx‐guided treatment on disease outcomes. The relationship of SLCO1B1 genotype and simvastatin acid systemic exposure in adults vs. children is a salient example. The impact of SLCO1B1 genotype is greater than twofold higher in children compared with that reported in adults. 34 Interestingly, although this was appreciated for simvastatin, but no appreciable differences were evident for its class‐mate pravastatin underscoring the importance of specific gene‐drug inquiries. 35 Careful consideration regarding measures of efficacy, tolerability, and drug retention is imperative to produce evidence that is translatable to the bedside. Study limitations Advantages of this study include our focus on a pediatric population and correlation of PGx testing results with the frequency of diseases and medication use among the patients tested. However, this study has several limitations. We obtained our sample from a group of patients referred to an academic children’s hospital that may have a higher burden of disease and thus may not represent the larger population of patients in the community. However, the prevalence of actionable genetic variants in our population mirrors that of adult literature, decreasing this concern about our sample. 16 The results from our study may not apply to different pediatric healthcare systems with different specialty clinics or referral patterns. We only examined guidance that contained genotypes included on the commercial PGx panel used at our institution. A panel that assessed different genes or a larger variety of genotypes would produce different results and might demonstrate a greater benefit of PGx in pediatric patients. Our findings regarding diagnoses and prescriptions were limited to those contained within our hospital’s EHRs. Diagnoses and prescriptions not documented in the EHR would not be captured, and neither would medications for indications that are not FDA‐approved or present in the Lexicomp. Therefore, our findings may underestimate the prevalence of relevant diagnoses and medication use in this population. In addition, we did not include medications used to treat the symptoms of acute pain and severe nausea (oxycodone, codeine, and ondansetron) in our analytic strategy. However, we estimated how many patients may benefit per year from PGx prior to using these medicines by examining the number of prescriptions for these medications and rates of actionable gene‐drug pairs in our sample. It remains unknown though how many of these prescriptions were first‐time prescriptions and how many were follow‐ups of previous prescriptions. Patients who had taken these medicines previously and found them effective and without side effects would be at much lower risk of having an underlying actionable PGx variant. Therefore, our method of estimation may overestimate the benefit of PGx testing associated with these medications. Given the retrospective nature of this study and limitations related to available clinical documentation within the EHR, we were unable to determine the impact of PGx testing on provider and family decision making. Future studies in pediatrics may consider exploring patients’ health care and medication use before and after testing, association of test results with patients’ experience of treatment efficacy and side effects, providers’ understanding of the PGx test results, and the influence of dosing guidelines on provider dosing practices. CONCLUSION Most children in our study had PGx variants that could impact their current treatment. Most of these acutely relevant findings were limited to three genes (e.g., CYP2D6, CYP2C19, and CYP3A5) and two major diagnosis groups (e.g., mental health disorders and gastritis/esophagitis/ulcer disease). Mental health disorders and gastritis/esophagitis/ulcer disease are prime targets for future study of PGx testing because of the high prevalence of these diagnoses and actionable gene‐drug‐diagnostic groups in children and adolescents. Considerations for future work also include the development of targeted pediatric PGx panels for dissemination in primary care that eliminate genes without any evidenced‐based guidelines or recommendations for drug management, for example, COMT, DRD2, GRIK4, HTR2A, HTR2C, IL28B, OPRM1, and SLC6A4, or are associated with drugs rarely used in children and adolescents (such as IFNL4‐peginterferon alfa‐2b). Limiting the number of genes tested may reduce complexity for the general practitioner in interpreting/returning results to patients and families. Development of a targeted pediatric PGx panel should ideally be informed by a cost:benefit analysis when determining which evidence‐based genes to omit (e.g., cost of repeat PGx testing at a later date and parental anxiety) balanced with the benefits of pre‐emptive testing to assist with diseases that might occur 40–50 years in the future. Through future study of the impact of PGx testing on patient outcomes and the optimal delivery of PGx findings to patients and families, we will learn how best to use this important tool to implement and practice precision medicine in pediatric patients. Funding S.S. is supported by a grant from the Eunice Kennedy Shriver National Institute for Child Health and Human Development T32 ND069038. Conflict of Interest T.R. has an investigator‐directed research grant from Merck to study contraception use among active duty servicewomen. All other authors declared no competing interests for this work. Author Contributions T.R., S.S., J.W., T.S., A.G., and B.B. wrote the manuscript. T.R. and S.S. designed the research. T.R. and S.S. performed the research. T.R. and S.S. analyzed the data. Supporting information Table S1 Click here for additional data file.
ARIPIPRAZOLE
DrugsGivenReaction
CC BY-NC
33048453
19,917,576
2021-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Ascites'.
The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis. Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKIs) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); median age 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12/15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients 5 could re-escalate the TKI dose and 9 improved their response. None of the 6 patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted. pmcIntroduction Tyrosine kinase inhibitors (TKI) are standard therapy for chronic myeloid leukemia (CML) and myelofibrosis (MF). Five TKI are currently approved for the treatment of CML in various stages, namely imatinib, nilotinib, dasatinib, bosutinib and ponatinib. Although these agents are generally well tolerated, some patients may develop adverse events, with myelosuppression being the most prominent.1-6 In most instances myelosuppression is grade 1 or 2 and requires no intervention. However, grade ≥3 thrombocytopenia (platelet ≤50x109/L) has been reported in 20% to 50% of patients. When this occurs, patients are usually managed with treatment interruption until platelets recovery (e.g., above 75x109/L) and dose reductions if thrombocytopenia recurs. Ruxolitinib is a JAK2 inhibitor used to manage splenomegaly and diseaseassociated symptoms in patients with MF.7 The dose limiting toxicity of ruxolitinib was thrombocytopenia8 and because of this the two pivotal phase III studies excluded patients with platelets ≤100x109/L. Still, thrombocytopenia was reported in 69% of patients, including 9% with grade ≥3.9 In patients with a platelet count of 50-100×109/L, grade ≥3 thrombocytopenia occurred in 56% of patients.10 Ruxolitinib-associated grade ≥3 thrombocytopenia is also typically managed with dose reductions or interruptions. Frequent dose interruptions and reductions might decrease TKI efficacy11,12 and may still not be sufficient to control thrombocytopenia. Patients who develop myelosuppression have a lower probability of achieving major or complete cytogenetic response (CCyR).11 For example, one study reported that patients treated with imatinib who developed grade ≥3 thrombocytopenia had a lower probability of CCyR compared to those who never developed thrombocytopenia (35% vs. 59%, P=0.02, respectively). Similarly, ruxolitinib efficacy is compromised with dose reductions and interruptions.12,13 In order to minimize dose reductions and interruptions, hematopoietic growth factors, filgrastim and erythropoietin stimulating agents (erythropoietin and darbepoetin) have been successfully used to manage neutropenia and anemia secondary to TKI in CML, respectively.14,15 Interleukin 11 (IL-11) was effective to manage thrombocytopenia associated with TKI in CML16 but use of this agent is associated with significant adverse events including fluid retention and cardiac arrhythmias. Eltrombopag is a non-peptide thrombopoietin receptor agonist that is effective and well tolerated among patients with immune thrombocytopenia, chronic hepatitis C-associated thrombocytopenia and severe aplastic anemia.17-19 It has also been safely used in acute myeloid leukemia without evidence of disease progression secondary to eltrombopag. 20 Here, we report the results from a pilot trial investigating the use of eltrombopag in the management of TKIor ruxolitinib-associated thrombocytopenia among patients with CML and MF. Methods Patients We conducted an open-label, non-randomized, phase II study of individualized dosing of eltrombopag. Eligible patients were aged 18 years or older with chronic phase CML receiving treatment with any Food and Drug Administration-approved TKI and experiencing grade ≥3 thrombocytopenia (platelets ≤50x109/L), or with MF receiving ruxolitinib and with platelets <100x109/L (since it is a dose-limiting toxicity and a label threshold for ruxolitinib), in either case after the first 3 months of therapy. Thrombocytopenia should have been either recurrent (i.e., be at least the second episode of thrombocytopenia) or have necessitated dose reductions of the TKI or ruxolitinib. All patients had signed an informed consent form approved by the Institutional Review Board, and the study was conducted in accordance with the Declaration of Helsinki. Study design Eltrombopag was commenced at 50 mg with dose escalation allowed every 2 weeks to 100 mg, 150 mg, 200 mg, and 300 mg (a higher dose than per label considering the thrombocytopenia refractoriness on these patients and the intent to continue TKI/ruxolitinib) according to platelet response. For patients of East Asian ancestry, eltrombopag was commenced at 25 mg daily with dose escalation allowed every 2 weeks. The following guideline was used to adjust dosing of eltrombopag: if the platelet count was >200x109/L at any time, the daily dose was reduced by 25 mg and re-assessed in 2 weeks; if >400x109/L, therapy was withheld and platelets assessed twice weekly until platelet count <150x109/L; therapy could then be resumed with the daily dose reduced by 25 mg. If the platelet count >400x109/L after 2 weeks was at the lowest dose, therapy was permanently discontinued. TKI doses were adjusted at the discretion of the treating physician per standard practice. Liver function tests (LFT) (alanine aminotransferase [ALT], aspartate aminotransferase [AST], and bilirubin) were done before the initiation of eltrombopag, every 2 weeks during the dose adjustment phase and following the monthly establishment of a stable dose. When LFT abnormalities were identified, LFT were performed weekly until the abnormalities resolved or stabilized; if ALT/AST levels were ≥ three-times the upper limit of normal (ULN): therapy was withheld, we then repeated abnormal liver function tests within 3-5 days; if confirmed abnormal, we monitored LFT weekly until resolved, stabilized, or returned to baseline. If ALT/AST levels ≥ three-times the ULN and were progressive, persistent (≥4 weeks), accompanied by increased direct bilirubin, or accompanied by clinical signs of liver injury or evidence of hepatic decompensation, eltrombopag was permanently discontinued. Patients who experienced other clinically significant grade 3 or greater toxicity possibly related to eltrombopag, had eltrombopag interruption until toxicity resolved to grade 1 or less. Treatment then was resumed at the immediate lower dose level. Failure to achieve a platelet count ≥50x109/L or ≥100x109/L in CML and MF patients, respectively after 8 weeks of eltrombopag was considered as lack of response. Statistical analysis Simon’s optimal two-stage design (Simon, 1989) was used to test the null hypothesis that the proportion of subjects with complete response is ≤0.10 versus the alternative that it is ≥0.30 (i.e., Po≤0.10 vs. Pa≥0.30) at alpha=0.05 with 80% power. The design resulted in an expected sample size of 15 and a probability of early termination of 0.736. The study was designed to study eltrombopag in ten patients in the first stage; the trial would be terminated if one or fewer achieved complete platelet response. Otherwise, the trial would go to the second stage, and 29 patients would be studied. If the total number of patients with complete platelet response were less than or equal to five, the drug would be deemed ineffective. The MF group was an exploratory group of ten patients to study the safety and activity of eltrombopag in patients with MF treated with ruxolitinib. We considered the activity promising if three or more patients out of ten achieved complete platelet response. For safety monitoring in the cohort with MF, accrual would stop if, at any time, four of ten patients encounter grade 3 or more nonhematological toxicity or progression to acute leukemia. As an additional safety procedure, we observed the first three MF patients on trial for at least 3 months before other patients were accrued. Response definitions Complete platelet response was defined as platelet count ≥50x109/L for CML, and ≥100x109/L for MF that was sustained for ≥3 months while continuing TKI or ruxolitinib therapy or with sustained (≥3 months) re-escalation of TKI dose without recurrence of thrombocytopenia. Criteria for CML and MF response were previously defined.21,22 The target response was a complete response in at least 30% of patients. Results Twenty-one patients were enrolled: 15 with CML and six with MF. Their median age was 60 years (range, 31-97) and their clinical characteristics are shown in Table 1. Median duration of disease was 2.2 years (range, 0.5-29 years) for patients with CML and 2 years (range, 0.3-3.6 years) for patients with MF. At the time of enrollment, patients with CML were receiving the following TKI: dasatinib (n=5), ponatinib (n=4), nilotinib (n=3), bosutinib (n=2), and imatinib (n=1), 27% were receiving their first TKI, 27% the second TKI, 27% the third, and 19% the fourth or later TKI. The median platelet count was 44x109/L (range, 3-49x109/L) in patients with CML and 62x109/L (range, 21-75x109/L) in those with MF. Cytogenetic response for patients with CML at baseline were partial in three, minor in six, and none in six. Prior therapies in MF patients were an investigational JAK2 inhibitor, and interferon a-2 in one patient each. The median dose of ruxolitinib was 10 mg (range, 10-30 mg) (Table 1). Eltrombopag dose distribution is summarized in Table 2. After a median duration of treatment of 18 months (range, 5-77 months), 12 of the 15 (80%) patients with CML achieved a complete platelet response with doses of eltrombopag of 50–300 mg per day. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). The median time to best response was 6 months (range, 2.1-13 months). Ten patients had sustained platelet recovery after stopping eltrombopag. The median duration for sustained platelet response was 45 months (range, 3-69 months). The three patients who did not achieve a complete platelet response had only minor changes in platelet count while they were taking eltrombopag (from 3x109/L to 8x109/L, 19x109/L to 45x109/L, and from 42x109/L to 46x109/L, respectively). Two patients (one each of CML and MF) had improvement in hemoglobin of over 2 g/dL from baseline (from 8.2 g/dL to 10.6 g/dL, and from 9.4 g/dL to 11.4 g/dL, respectively), Hemoglobin improvement was sustained over 21.5 and 2 months respectively while patients were taking eltrombopag. Hemoglobin levels declined after interruption of eltrombopag. One patient with CML had an absolute neutrophil count recovery to >1x109/L (baseline neutrophils 0.71x109/L). Absolute neutrophil count improvement was sustained for >6 months while on eltrombopag. Absolute neutrophil count then declined after interruption of eltrombopag. The TKI doses and duration for patients with CML post enrollment are summarized in Table 1. Nine patients with CML experienced an improvement in the cytogenetic response during the observation period (all of them had sustained platelet recovery after stopping eltrombopag); one from none to complete, two from minor to complete, four from minor to partial, and two from partial to complete (Table 3). In five patients with CML the TKI dose was increased and maintained while continuing eltrombopag. Dasatinib daily dose was increased from 50 mg to 100 mg in three patients, nilotinib dose was increased in one patient form 150 mg twice daily to 200 mg twice daily, and one patient had an increase in ponatinib dose from 15 mg every other day to 15 mg daily. There were no TKI dose-limiting toxicities in patients who increased their TKI doses. The dose increase was associated with improvement in CML response in four of these five patients. In the five CML patients who had a cytogenetic response but did not have TKI dose escalation, the improvement in cytogenetic response was noticed while patients were on eltrombopag. Three CML patients had a switch in their TKI (Online Supplementary Table S1). All three of these patients had already some improvement in thrombocytopenia before switching their TKI, with the change indicated for other non-hematologic adverse events in one patient and the inefficacy of the TKI in the other two patients. None of the six patients with MF responded (i.e., none had a sustained increase in platelet count to ≥100x109/L); minor upward transient variations in platelet counts were seen in three patients (from 21x109/L to 28x109/L, 41x109/L to 55x109/L and from 65x109L to 75x109/L, respectively). Table 1. Baseline characteristics. Table 2. Eltrombopag dose distribution, mg per day (all patients). As of the date of this report, 20 patients were off study because of a lack of response (n=9), stem cell transplant (n=2), death (n=2), patient’s wish (n=1), adverse events (n=2), TKI discontinuation (n=1), loss to follow-up (n=1) and stable platelets (n=2). The two deaths on study were not related to treatment. One death was secondary to infectious complication in a patient with MF. The second death was secondary to hemorrhagic shock in a CML patient. This patient, treated with dasatinib, developed hepatosplenomegaly and ascites while on study but the etiology was not known. There was no evidence of portal vein thrombosis on CT abdomen/pelvis. Both eltrombopag and dasatinib were held and she had no platelets response. The platelet count was 10x109/L at the time of death due to severe gastrointestinal and genitourinary bleeding. Figure 1. Platelet change from baseline to response. *Each blue bar reflects change in platelet count in a chronic myeloid leukemia patient, while each green bar reflects change in platelet count in a myelofibrosis patient. Therapy was well tolerated in most patients, but two patients on ponatinib developed thrombotic events. Two months after eltrombopag discontinuation due to termination of the study, one patient with CML developed significant thrombocytosis (>1,000x109/L) with a white blood cell count of 9.5x109/L, 3% basophils, and 2% peripheral blast accompanied by headache and eye pain. Ophthalmoscopic examination was suggestive of bilateral plaques or thrombosis in the retinal vasculature but fluoroscopic evaluation did not reveal retinal vasculature blockage. Ponatinib was discontinued and thrombocytosis was managed with hydroxyurea. The aforementioned symptoms resolved. There was no cytogenetic response prior or after starting eltrombopag. Seven months after stopping eltrombopag, the patient had a persistent increase in blasts to 13% without a lack of hematologic response and she was then started on a clinical trial with an investigational TKI. Another CML patient developed non-occlusive deep venous thrombosis in the right upper extremity without thrombocytosis while on ponatinib 4 months after the study was terminated. One MF patient who had a history of coronary artery disease status post coronary artery bypass surgery developed myocardial infarction (MI) while on eltrombopag. This patent had then worsening increase in bone marrow fibrosis from grade 2 to grade 3 and was taken off study 40 days after MI. No further additional thrombotic/thromboembolic complications in CML and MF patients observed during or after the study (Online Supplementary Table S2). One patient (CML) discontinued therapy due to toxicity (elevation of liver function tests). Grade 3/4 toxicities irrespective of attribution listed in Table 4. One patient with MF had an increase in bone marrow fibrosis from grade 2 to grade 3. That patient had an increase in blast from 3% to 8% in the peripheral blood and an increase from 1% to 6% in the bone marrow while he was on study but with an improvement in hemoglobin. There was no change in the patient's disease other than this change in blast percentage. The patient was taken off study for lack of platelet response and later started on another clinical trial (PRM-151 + ruxolitinib). No progression of disease has been documented in any other patients. No clonal evolution was observed in patients with prolonged eltrombopag use. Discussion Thrombocytopenia is a common adverse event in patients with CML and MF who are treated with TKI and ruxolitinib, respectively.10,23 In most instances, thrombocytopenia is transient, occurs early during treatment initiation, and can be successfully managed with transient treatment interruptions and occasionally dose adjustments. However, in some patients thrombocytopenia can be persistent and more severe requiring frequent treatment interruptions and dose reductions, which might adversely influence treatment outcome.11 To that end, rIL-11 was successfully used in CML patients for the management of TKI associated thrombocytopenia.16 The main limitation of use of rIL-11 in the management of chemotherapy-induced thrombocytopenia in solid malignancies was the narrow therapeutic window with significant fluid retention and occasional arrhythmias. However, at lower doses used in CML, it was well tolerated24,25 with grade 1 or 2 peripheral edema observed in six patients (43%). Eltrombopag is a second generation oral thrombopoietin receptor agonist that has induced improvement of thrombocytopenia in patients with immune-mediate thrombocytopenia (ITP) or aplastic anemia. The EXTEND trial demonstrated that long-term use of eltrombopag was effective in maintaining for more than 6 months platelet counts of 50×109/L or more and reducing bleeding in most patients with ITP. Addition of eltrombopag to immunosuppressive treatment also markedly increased overall and complete hematologic response rates in treatment-naive severe aplastic anemia.26 Here we describe the use of eltrombopag in the management of TKI-related thrombocytopenia in CML and MF. Our results suggest clinical benefit in most patients with CML with a generally favorable safety profile, although two patents (both on ponartinib) had thrombotic events. In contrast, no response was observed in patients with MF. Theoretical concerns about the use of eltrombopag in this setting include increase in marrow blasts and possible transformation to advanced phases, thrombotic events including portal vein thrombosis, and increase in marrow fibrosis. We did not observe any instance of transformation in our series, in concordance with pre-clinical and clinical data showing no evidence of worsening leukemia.20,27 There was also no increase in marrow fibrosis in CML patients. Our series is small so the lack of such events should be considered as preliminary but reassuring. The most common adverse event was LFT elevation, but these were generally transient, reversible and manageable with dose adjustments. However, in one case it led to discontinuation of eltrombopag because of recurrent transaminitis. Two patients who received ponatinib (50%) had thrombotic events while on eltrombopag, this might raise the precaution of using ponatinib in conjunction with eltrombopag in CML patients. Despite the median disease duration of 2.2 years and multiple TKI use in CML patients before enrollment, eltrombopag demonstrated clinical efficacy with complete platelet response of 80% (12 of 15). This compares favorably to what was reported with rIL-11.16 More important, nine patients (60%) had improvement in cytogenetic responses, likely the result of a more sustained therapy with TKI. Notably, as doses of eltrombopag were increased, LFT elevations were noted in some patients. Conversely, eltrombopag dose interruptions or reductions due to such events or to platelets reaching >200x109/L, occasionally resulted in a drop-in platelet counts. Thus, close monitoring and dynamic management is required, at least during the initial stages of therapy to obtain the maximum effect while maintaining safety. The lack of efficacy among patients with MF could be in part secondary to advanced disease, or possible antagonism between the two medications. Thrombopoietin agonist are dependent on JAK-stat pathway which is inhibited by ruxolitinib.28 Table 3. Response to eltrombopag in chronic myeloid leukemia patients. Table 4. Treatment emergent adverse events. Our study has several limitations. It was a small study, and it did not accrue to the target sample size of 29 patients due to slow enrollment making the observation preliminary and requiring confirmation. We also do not have evidence or investigation of any immune mechanisms associated with thrombocytopenia, although we believe it is unlikely that these patients with CML had an immune mediated thrombocytopenia, and uncommon occurrence in this setting. In conclusion, our findings show that eltrombopag doses up to 300 mg may alleviate TKI-associated thrombocytopenia in some patients with CML. No similar benefit has been observed in patients with MF treated with ruxolitinib. Although generally safe, thrombotic events were noted that deserve further investigation, particularly when used in combination with ponatinib. Additional studies are warranted to confirm these observations. Supplementary Material Supplementary Appendix
DASATINIB
DrugsGivenReaction
CC BY-NC
33054123
20,176,531
2021-11-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Gastrointestinal haemorrhage'.
The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis. Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKIs) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); median age 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12/15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients 5 could re-escalate the TKI dose and 9 improved their response. None of the 6 patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted. pmcIntroduction Tyrosine kinase inhibitors (TKI) are standard therapy for chronic myeloid leukemia (CML) and myelofibrosis (MF). Five TKI are currently approved for the treatment of CML in various stages, namely imatinib, nilotinib, dasatinib, bosutinib and ponatinib. Although these agents are generally well tolerated, some patients may develop adverse events, with myelosuppression being the most prominent.1-6 In most instances myelosuppression is grade 1 or 2 and requires no intervention. However, grade ≥3 thrombocytopenia (platelet ≤50x109/L) has been reported in 20% to 50% of patients. When this occurs, patients are usually managed with treatment interruption until platelets recovery (e.g., above 75x109/L) and dose reductions if thrombocytopenia recurs. Ruxolitinib is a JAK2 inhibitor used to manage splenomegaly and diseaseassociated symptoms in patients with MF.7 The dose limiting toxicity of ruxolitinib was thrombocytopenia8 and because of this the two pivotal phase III studies excluded patients with platelets ≤100x109/L. Still, thrombocytopenia was reported in 69% of patients, including 9% with grade ≥3.9 In patients with a platelet count of 50-100×109/L, grade ≥3 thrombocytopenia occurred in 56% of patients.10 Ruxolitinib-associated grade ≥3 thrombocytopenia is also typically managed with dose reductions or interruptions. Frequent dose interruptions and reductions might decrease TKI efficacy11,12 and may still not be sufficient to control thrombocytopenia. Patients who develop myelosuppression have a lower probability of achieving major or complete cytogenetic response (CCyR).11 For example, one study reported that patients treated with imatinib who developed grade ≥3 thrombocytopenia had a lower probability of CCyR compared to those who never developed thrombocytopenia (35% vs. 59%, P=0.02, respectively). Similarly, ruxolitinib efficacy is compromised with dose reductions and interruptions.12,13 In order to minimize dose reductions and interruptions, hematopoietic growth factors, filgrastim and erythropoietin stimulating agents (erythropoietin and darbepoetin) have been successfully used to manage neutropenia and anemia secondary to TKI in CML, respectively.14,15 Interleukin 11 (IL-11) was effective to manage thrombocytopenia associated with TKI in CML16 but use of this agent is associated with significant adverse events including fluid retention and cardiac arrhythmias. Eltrombopag is a non-peptide thrombopoietin receptor agonist that is effective and well tolerated among patients with immune thrombocytopenia, chronic hepatitis C-associated thrombocytopenia and severe aplastic anemia.17-19 It has also been safely used in acute myeloid leukemia without evidence of disease progression secondary to eltrombopag. 20 Here, we report the results from a pilot trial investigating the use of eltrombopag in the management of TKIor ruxolitinib-associated thrombocytopenia among patients with CML and MF. Methods Patients We conducted an open-label, non-randomized, phase II study of individualized dosing of eltrombopag. Eligible patients were aged 18 years or older with chronic phase CML receiving treatment with any Food and Drug Administration-approved TKI and experiencing grade ≥3 thrombocytopenia (platelets ≤50x109/L), or with MF receiving ruxolitinib and with platelets <100x109/L (since it is a dose-limiting toxicity and a label threshold for ruxolitinib), in either case after the first 3 months of therapy. Thrombocytopenia should have been either recurrent (i.e., be at least the second episode of thrombocytopenia) or have necessitated dose reductions of the TKI or ruxolitinib. All patients had signed an informed consent form approved by the Institutional Review Board, and the study was conducted in accordance with the Declaration of Helsinki. Study design Eltrombopag was commenced at 50 mg with dose escalation allowed every 2 weeks to 100 mg, 150 mg, 200 mg, and 300 mg (a higher dose than per label considering the thrombocytopenia refractoriness on these patients and the intent to continue TKI/ruxolitinib) according to platelet response. For patients of East Asian ancestry, eltrombopag was commenced at 25 mg daily with dose escalation allowed every 2 weeks. The following guideline was used to adjust dosing of eltrombopag: if the platelet count was >200x109/L at any time, the daily dose was reduced by 25 mg and re-assessed in 2 weeks; if >400x109/L, therapy was withheld and platelets assessed twice weekly until platelet count <150x109/L; therapy could then be resumed with the daily dose reduced by 25 mg. If the platelet count >400x109/L after 2 weeks was at the lowest dose, therapy was permanently discontinued. TKI doses were adjusted at the discretion of the treating physician per standard practice. Liver function tests (LFT) (alanine aminotransferase [ALT], aspartate aminotransferase [AST], and bilirubin) were done before the initiation of eltrombopag, every 2 weeks during the dose adjustment phase and following the monthly establishment of a stable dose. When LFT abnormalities were identified, LFT were performed weekly until the abnormalities resolved or stabilized; if ALT/AST levels were ≥ three-times the upper limit of normal (ULN): therapy was withheld, we then repeated abnormal liver function tests within 3-5 days; if confirmed abnormal, we monitored LFT weekly until resolved, stabilized, or returned to baseline. If ALT/AST levels ≥ three-times the ULN and were progressive, persistent (≥4 weeks), accompanied by increased direct bilirubin, or accompanied by clinical signs of liver injury or evidence of hepatic decompensation, eltrombopag was permanently discontinued. Patients who experienced other clinically significant grade 3 or greater toxicity possibly related to eltrombopag, had eltrombopag interruption until toxicity resolved to grade 1 or less. Treatment then was resumed at the immediate lower dose level. Failure to achieve a platelet count ≥50x109/L or ≥100x109/L in CML and MF patients, respectively after 8 weeks of eltrombopag was considered as lack of response. Statistical analysis Simon’s optimal two-stage design (Simon, 1989) was used to test the null hypothesis that the proportion of subjects with complete response is ≤0.10 versus the alternative that it is ≥0.30 (i.e., Po≤0.10 vs. Pa≥0.30) at alpha=0.05 with 80% power. The design resulted in an expected sample size of 15 and a probability of early termination of 0.736. The study was designed to study eltrombopag in ten patients in the first stage; the trial would be terminated if one or fewer achieved complete platelet response. Otherwise, the trial would go to the second stage, and 29 patients would be studied. If the total number of patients with complete platelet response were less than or equal to five, the drug would be deemed ineffective. The MF group was an exploratory group of ten patients to study the safety and activity of eltrombopag in patients with MF treated with ruxolitinib. We considered the activity promising if three or more patients out of ten achieved complete platelet response. For safety monitoring in the cohort with MF, accrual would stop if, at any time, four of ten patients encounter grade 3 or more nonhematological toxicity or progression to acute leukemia. As an additional safety procedure, we observed the first three MF patients on trial for at least 3 months before other patients were accrued. Response definitions Complete platelet response was defined as platelet count ≥50x109/L for CML, and ≥100x109/L for MF that was sustained for ≥3 months while continuing TKI or ruxolitinib therapy or with sustained (≥3 months) re-escalation of TKI dose without recurrence of thrombocytopenia. Criteria for CML and MF response were previously defined.21,22 The target response was a complete response in at least 30% of patients. Results Twenty-one patients were enrolled: 15 with CML and six with MF. Their median age was 60 years (range, 31-97) and their clinical characteristics are shown in Table 1. Median duration of disease was 2.2 years (range, 0.5-29 years) for patients with CML and 2 years (range, 0.3-3.6 years) for patients with MF. At the time of enrollment, patients with CML were receiving the following TKI: dasatinib (n=5), ponatinib (n=4), nilotinib (n=3), bosutinib (n=2), and imatinib (n=1), 27% were receiving their first TKI, 27% the second TKI, 27% the third, and 19% the fourth or later TKI. The median platelet count was 44x109/L (range, 3-49x109/L) in patients with CML and 62x109/L (range, 21-75x109/L) in those with MF. Cytogenetic response for patients with CML at baseline were partial in three, minor in six, and none in six. Prior therapies in MF patients were an investigational JAK2 inhibitor, and interferon a-2 in one patient each. The median dose of ruxolitinib was 10 mg (range, 10-30 mg) (Table 1). Eltrombopag dose distribution is summarized in Table 2. After a median duration of treatment of 18 months (range, 5-77 months), 12 of the 15 (80%) patients with CML achieved a complete platelet response with doses of eltrombopag of 50–300 mg per day. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). The median time to best response was 6 months (range, 2.1-13 months). Ten patients had sustained platelet recovery after stopping eltrombopag. The median duration for sustained platelet response was 45 months (range, 3-69 months). The three patients who did not achieve a complete platelet response had only minor changes in platelet count while they were taking eltrombopag (from 3x109/L to 8x109/L, 19x109/L to 45x109/L, and from 42x109/L to 46x109/L, respectively). Two patients (one each of CML and MF) had improvement in hemoglobin of over 2 g/dL from baseline (from 8.2 g/dL to 10.6 g/dL, and from 9.4 g/dL to 11.4 g/dL, respectively), Hemoglobin improvement was sustained over 21.5 and 2 months respectively while patients were taking eltrombopag. Hemoglobin levels declined after interruption of eltrombopag. One patient with CML had an absolute neutrophil count recovery to >1x109/L (baseline neutrophils 0.71x109/L). Absolute neutrophil count improvement was sustained for >6 months while on eltrombopag. Absolute neutrophil count then declined after interruption of eltrombopag. The TKI doses and duration for patients with CML post enrollment are summarized in Table 1. Nine patients with CML experienced an improvement in the cytogenetic response during the observation period (all of them had sustained platelet recovery after stopping eltrombopag); one from none to complete, two from minor to complete, four from minor to partial, and two from partial to complete (Table 3). In five patients with CML the TKI dose was increased and maintained while continuing eltrombopag. Dasatinib daily dose was increased from 50 mg to 100 mg in three patients, nilotinib dose was increased in one patient form 150 mg twice daily to 200 mg twice daily, and one patient had an increase in ponatinib dose from 15 mg every other day to 15 mg daily. There were no TKI dose-limiting toxicities in patients who increased their TKI doses. The dose increase was associated with improvement in CML response in four of these five patients. In the five CML patients who had a cytogenetic response but did not have TKI dose escalation, the improvement in cytogenetic response was noticed while patients were on eltrombopag. Three CML patients had a switch in their TKI (Online Supplementary Table S1). All three of these patients had already some improvement in thrombocytopenia before switching their TKI, with the change indicated for other non-hematologic adverse events in one patient and the inefficacy of the TKI in the other two patients. None of the six patients with MF responded (i.e., none had a sustained increase in platelet count to ≥100x109/L); minor upward transient variations in platelet counts were seen in three patients (from 21x109/L to 28x109/L, 41x109/L to 55x109/L and from 65x109L to 75x109/L, respectively). Table 1. Baseline characteristics. Table 2. Eltrombopag dose distribution, mg per day (all patients). As of the date of this report, 20 patients were off study because of a lack of response (n=9), stem cell transplant (n=2), death (n=2), patient’s wish (n=1), adverse events (n=2), TKI discontinuation (n=1), loss to follow-up (n=1) and stable platelets (n=2). The two deaths on study were not related to treatment. One death was secondary to infectious complication in a patient with MF. The second death was secondary to hemorrhagic shock in a CML patient. This patient, treated with dasatinib, developed hepatosplenomegaly and ascites while on study but the etiology was not known. There was no evidence of portal vein thrombosis on CT abdomen/pelvis. Both eltrombopag and dasatinib were held and she had no platelets response. The platelet count was 10x109/L at the time of death due to severe gastrointestinal and genitourinary bleeding. Figure 1. Platelet change from baseline to response. *Each blue bar reflects change in platelet count in a chronic myeloid leukemia patient, while each green bar reflects change in platelet count in a myelofibrosis patient. Therapy was well tolerated in most patients, but two patients on ponatinib developed thrombotic events. Two months after eltrombopag discontinuation due to termination of the study, one patient with CML developed significant thrombocytosis (>1,000x109/L) with a white blood cell count of 9.5x109/L, 3% basophils, and 2% peripheral blast accompanied by headache and eye pain. Ophthalmoscopic examination was suggestive of bilateral plaques or thrombosis in the retinal vasculature but fluoroscopic evaluation did not reveal retinal vasculature blockage. Ponatinib was discontinued and thrombocytosis was managed with hydroxyurea. The aforementioned symptoms resolved. There was no cytogenetic response prior or after starting eltrombopag. Seven months after stopping eltrombopag, the patient had a persistent increase in blasts to 13% without a lack of hematologic response and she was then started on a clinical trial with an investigational TKI. Another CML patient developed non-occlusive deep venous thrombosis in the right upper extremity without thrombocytosis while on ponatinib 4 months after the study was terminated. One MF patient who had a history of coronary artery disease status post coronary artery bypass surgery developed myocardial infarction (MI) while on eltrombopag. This patent had then worsening increase in bone marrow fibrosis from grade 2 to grade 3 and was taken off study 40 days after MI. No further additional thrombotic/thromboembolic complications in CML and MF patients observed during or after the study (Online Supplementary Table S2). One patient (CML) discontinued therapy due to toxicity (elevation of liver function tests). Grade 3/4 toxicities irrespective of attribution listed in Table 4. One patient with MF had an increase in bone marrow fibrosis from grade 2 to grade 3. That patient had an increase in blast from 3% to 8% in the peripheral blood and an increase from 1% to 6% in the bone marrow while he was on study but with an improvement in hemoglobin. There was no change in the patient's disease other than this change in blast percentage. The patient was taken off study for lack of platelet response and later started on another clinical trial (PRM-151 + ruxolitinib). No progression of disease has been documented in any other patients. No clonal evolution was observed in patients with prolonged eltrombopag use. Discussion Thrombocytopenia is a common adverse event in patients with CML and MF who are treated with TKI and ruxolitinib, respectively.10,23 In most instances, thrombocytopenia is transient, occurs early during treatment initiation, and can be successfully managed with transient treatment interruptions and occasionally dose adjustments. However, in some patients thrombocytopenia can be persistent and more severe requiring frequent treatment interruptions and dose reductions, which might adversely influence treatment outcome.11 To that end, rIL-11 was successfully used in CML patients for the management of TKI associated thrombocytopenia.16 The main limitation of use of rIL-11 in the management of chemotherapy-induced thrombocytopenia in solid malignancies was the narrow therapeutic window with significant fluid retention and occasional arrhythmias. However, at lower doses used in CML, it was well tolerated24,25 with grade 1 or 2 peripheral edema observed in six patients (43%). Eltrombopag is a second generation oral thrombopoietin receptor agonist that has induced improvement of thrombocytopenia in patients with immune-mediate thrombocytopenia (ITP) or aplastic anemia. The EXTEND trial demonstrated that long-term use of eltrombopag was effective in maintaining for more than 6 months platelet counts of 50×109/L or more and reducing bleeding in most patients with ITP. Addition of eltrombopag to immunosuppressive treatment also markedly increased overall and complete hematologic response rates in treatment-naive severe aplastic anemia.26 Here we describe the use of eltrombopag in the management of TKI-related thrombocytopenia in CML and MF. Our results suggest clinical benefit in most patients with CML with a generally favorable safety profile, although two patents (both on ponartinib) had thrombotic events. In contrast, no response was observed in patients with MF. Theoretical concerns about the use of eltrombopag in this setting include increase in marrow blasts and possible transformation to advanced phases, thrombotic events including portal vein thrombosis, and increase in marrow fibrosis. We did not observe any instance of transformation in our series, in concordance with pre-clinical and clinical data showing no evidence of worsening leukemia.20,27 There was also no increase in marrow fibrosis in CML patients. Our series is small so the lack of such events should be considered as preliminary but reassuring. The most common adverse event was LFT elevation, but these were generally transient, reversible and manageable with dose adjustments. However, in one case it led to discontinuation of eltrombopag because of recurrent transaminitis. Two patients who received ponatinib (50%) had thrombotic events while on eltrombopag, this might raise the precaution of using ponatinib in conjunction with eltrombopag in CML patients. Despite the median disease duration of 2.2 years and multiple TKI use in CML patients before enrollment, eltrombopag demonstrated clinical efficacy with complete platelet response of 80% (12 of 15). This compares favorably to what was reported with rIL-11.16 More important, nine patients (60%) had improvement in cytogenetic responses, likely the result of a more sustained therapy with TKI. Notably, as doses of eltrombopag were increased, LFT elevations were noted in some patients. Conversely, eltrombopag dose interruptions or reductions due to such events or to platelets reaching >200x109/L, occasionally resulted in a drop-in platelet counts. Thus, close monitoring and dynamic management is required, at least during the initial stages of therapy to obtain the maximum effect while maintaining safety. The lack of efficacy among patients with MF could be in part secondary to advanced disease, or possible antagonism between the two medications. Thrombopoietin agonist are dependent on JAK-stat pathway which is inhibited by ruxolitinib.28 Table 3. Response to eltrombopag in chronic myeloid leukemia patients. Table 4. Treatment emergent adverse events. Our study has several limitations. It was a small study, and it did not accrue to the target sample size of 29 patients due to slow enrollment making the observation preliminary and requiring confirmation. We also do not have evidence or investigation of any immune mechanisms associated with thrombocytopenia, although we believe it is unlikely that these patients with CML had an immune mediated thrombocytopenia, and uncommon occurrence in this setting. In conclusion, our findings show that eltrombopag doses up to 300 mg may alleviate TKI-associated thrombocytopenia in some patients with CML. No similar benefit has been observed in patients with MF treated with ruxolitinib. Although generally safe, thrombotic events were noted that deserve further investigation, particularly when used in combination with ponatinib. Additional studies are warranted to confirm these observations. Supplementary Material Supplementary Appendix
DASATINIB
DrugsGivenReaction
CC BY-NC
33054123
20,176,531
2021-11-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Hepatosplenomegaly'.
The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis. Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKIs) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); median age 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12/15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients 5 could re-escalate the TKI dose and 9 improved their response. None of the 6 patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted. pmcIntroduction Tyrosine kinase inhibitors (TKI) are standard therapy for chronic myeloid leukemia (CML) and myelofibrosis (MF). Five TKI are currently approved for the treatment of CML in various stages, namely imatinib, nilotinib, dasatinib, bosutinib and ponatinib. Although these agents are generally well tolerated, some patients may develop adverse events, with myelosuppression being the most prominent.1-6 In most instances myelosuppression is grade 1 or 2 and requires no intervention. However, grade ≥3 thrombocytopenia (platelet ≤50x109/L) has been reported in 20% to 50% of patients. When this occurs, patients are usually managed with treatment interruption until platelets recovery (e.g., above 75x109/L) and dose reductions if thrombocytopenia recurs. Ruxolitinib is a JAK2 inhibitor used to manage splenomegaly and diseaseassociated symptoms in patients with MF.7 The dose limiting toxicity of ruxolitinib was thrombocytopenia8 and because of this the two pivotal phase III studies excluded patients with platelets ≤100x109/L. Still, thrombocytopenia was reported in 69% of patients, including 9% with grade ≥3.9 In patients with a platelet count of 50-100×109/L, grade ≥3 thrombocytopenia occurred in 56% of patients.10 Ruxolitinib-associated grade ≥3 thrombocytopenia is also typically managed with dose reductions or interruptions. Frequent dose interruptions and reductions might decrease TKI efficacy11,12 and may still not be sufficient to control thrombocytopenia. Patients who develop myelosuppression have a lower probability of achieving major or complete cytogenetic response (CCyR).11 For example, one study reported that patients treated with imatinib who developed grade ≥3 thrombocytopenia had a lower probability of CCyR compared to those who never developed thrombocytopenia (35% vs. 59%, P=0.02, respectively). Similarly, ruxolitinib efficacy is compromised with dose reductions and interruptions.12,13 In order to minimize dose reductions and interruptions, hematopoietic growth factors, filgrastim and erythropoietin stimulating agents (erythropoietin and darbepoetin) have been successfully used to manage neutropenia and anemia secondary to TKI in CML, respectively.14,15 Interleukin 11 (IL-11) was effective to manage thrombocytopenia associated with TKI in CML16 but use of this agent is associated with significant adverse events including fluid retention and cardiac arrhythmias. Eltrombopag is a non-peptide thrombopoietin receptor agonist that is effective and well tolerated among patients with immune thrombocytopenia, chronic hepatitis C-associated thrombocytopenia and severe aplastic anemia.17-19 It has also been safely used in acute myeloid leukemia without evidence of disease progression secondary to eltrombopag. 20 Here, we report the results from a pilot trial investigating the use of eltrombopag in the management of TKIor ruxolitinib-associated thrombocytopenia among patients with CML and MF. Methods Patients We conducted an open-label, non-randomized, phase II study of individualized dosing of eltrombopag. Eligible patients were aged 18 years or older with chronic phase CML receiving treatment with any Food and Drug Administration-approved TKI and experiencing grade ≥3 thrombocytopenia (platelets ≤50x109/L), or with MF receiving ruxolitinib and with platelets <100x109/L (since it is a dose-limiting toxicity and a label threshold for ruxolitinib), in either case after the first 3 months of therapy. Thrombocytopenia should have been either recurrent (i.e., be at least the second episode of thrombocytopenia) or have necessitated dose reductions of the TKI or ruxolitinib. All patients had signed an informed consent form approved by the Institutional Review Board, and the study was conducted in accordance with the Declaration of Helsinki. Study design Eltrombopag was commenced at 50 mg with dose escalation allowed every 2 weeks to 100 mg, 150 mg, 200 mg, and 300 mg (a higher dose than per label considering the thrombocytopenia refractoriness on these patients and the intent to continue TKI/ruxolitinib) according to platelet response. For patients of East Asian ancestry, eltrombopag was commenced at 25 mg daily with dose escalation allowed every 2 weeks. The following guideline was used to adjust dosing of eltrombopag: if the platelet count was >200x109/L at any time, the daily dose was reduced by 25 mg and re-assessed in 2 weeks; if >400x109/L, therapy was withheld and platelets assessed twice weekly until platelet count <150x109/L; therapy could then be resumed with the daily dose reduced by 25 mg. If the platelet count >400x109/L after 2 weeks was at the lowest dose, therapy was permanently discontinued. TKI doses were adjusted at the discretion of the treating physician per standard practice. Liver function tests (LFT) (alanine aminotransferase [ALT], aspartate aminotransferase [AST], and bilirubin) were done before the initiation of eltrombopag, every 2 weeks during the dose adjustment phase and following the monthly establishment of a stable dose. When LFT abnormalities were identified, LFT were performed weekly until the abnormalities resolved or stabilized; if ALT/AST levels were ≥ three-times the upper limit of normal (ULN): therapy was withheld, we then repeated abnormal liver function tests within 3-5 days; if confirmed abnormal, we monitored LFT weekly until resolved, stabilized, or returned to baseline. If ALT/AST levels ≥ three-times the ULN and were progressive, persistent (≥4 weeks), accompanied by increased direct bilirubin, or accompanied by clinical signs of liver injury or evidence of hepatic decompensation, eltrombopag was permanently discontinued. Patients who experienced other clinically significant grade 3 or greater toxicity possibly related to eltrombopag, had eltrombopag interruption until toxicity resolved to grade 1 or less. Treatment then was resumed at the immediate lower dose level. Failure to achieve a platelet count ≥50x109/L or ≥100x109/L in CML and MF patients, respectively after 8 weeks of eltrombopag was considered as lack of response. Statistical analysis Simon’s optimal two-stage design (Simon, 1989) was used to test the null hypothesis that the proportion of subjects with complete response is ≤0.10 versus the alternative that it is ≥0.30 (i.e., Po≤0.10 vs. Pa≥0.30) at alpha=0.05 with 80% power. The design resulted in an expected sample size of 15 and a probability of early termination of 0.736. The study was designed to study eltrombopag in ten patients in the first stage; the trial would be terminated if one or fewer achieved complete platelet response. Otherwise, the trial would go to the second stage, and 29 patients would be studied. If the total number of patients with complete platelet response were less than or equal to five, the drug would be deemed ineffective. The MF group was an exploratory group of ten patients to study the safety and activity of eltrombopag in patients with MF treated with ruxolitinib. We considered the activity promising if three or more patients out of ten achieved complete platelet response. For safety monitoring in the cohort with MF, accrual would stop if, at any time, four of ten patients encounter grade 3 or more nonhematological toxicity or progression to acute leukemia. As an additional safety procedure, we observed the first three MF patients on trial for at least 3 months before other patients were accrued. Response definitions Complete platelet response was defined as platelet count ≥50x109/L for CML, and ≥100x109/L for MF that was sustained for ≥3 months while continuing TKI or ruxolitinib therapy or with sustained (≥3 months) re-escalation of TKI dose without recurrence of thrombocytopenia. Criteria for CML and MF response were previously defined.21,22 The target response was a complete response in at least 30% of patients. Results Twenty-one patients were enrolled: 15 with CML and six with MF. Their median age was 60 years (range, 31-97) and their clinical characteristics are shown in Table 1. Median duration of disease was 2.2 years (range, 0.5-29 years) for patients with CML and 2 years (range, 0.3-3.6 years) for patients with MF. At the time of enrollment, patients with CML were receiving the following TKI: dasatinib (n=5), ponatinib (n=4), nilotinib (n=3), bosutinib (n=2), and imatinib (n=1), 27% were receiving their first TKI, 27% the second TKI, 27% the third, and 19% the fourth or later TKI. The median platelet count was 44x109/L (range, 3-49x109/L) in patients with CML and 62x109/L (range, 21-75x109/L) in those with MF. Cytogenetic response for patients with CML at baseline were partial in three, minor in six, and none in six. Prior therapies in MF patients were an investigational JAK2 inhibitor, and interferon a-2 in one patient each. The median dose of ruxolitinib was 10 mg (range, 10-30 mg) (Table 1). Eltrombopag dose distribution is summarized in Table 2. After a median duration of treatment of 18 months (range, 5-77 months), 12 of the 15 (80%) patients with CML achieved a complete platelet response with doses of eltrombopag of 50–300 mg per day. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). The median time to best response was 6 months (range, 2.1-13 months). Ten patients had sustained platelet recovery after stopping eltrombopag. The median duration for sustained platelet response was 45 months (range, 3-69 months). The three patients who did not achieve a complete platelet response had only minor changes in platelet count while they were taking eltrombopag (from 3x109/L to 8x109/L, 19x109/L to 45x109/L, and from 42x109/L to 46x109/L, respectively). Two patients (one each of CML and MF) had improvement in hemoglobin of over 2 g/dL from baseline (from 8.2 g/dL to 10.6 g/dL, and from 9.4 g/dL to 11.4 g/dL, respectively), Hemoglobin improvement was sustained over 21.5 and 2 months respectively while patients were taking eltrombopag. Hemoglobin levels declined after interruption of eltrombopag. One patient with CML had an absolute neutrophil count recovery to >1x109/L (baseline neutrophils 0.71x109/L). Absolute neutrophil count improvement was sustained for >6 months while on eltrombopag. Absolute neutrophil count then declined after interruption of eltrombopag. The TKI doses and duration for patients with CML post enrollment are summarized in Table 1. Nine patients with CML experienced an improvement in the cytogenetic response during the observation period (all of them had sustained platelet recovery after stopping eltrombopag); one from none to complete, two from minor to complete, four from minor to partial, and two from partial to complete (Table 3). In five patients with CML the TKI dose was increased and maintained while continuing eltrombopag. Dasatinib daily dose was increased from 50 mg to 100 mg in three patients, nilotinib dose was increased in one patient form 150 mg twice daily to 200 mg twice daily, and one patient had an increase in ponatinib dose from 15 mg every other day to 15 mg daily. There were no TKI dose-limiting toxicities in patients who increased their TKI doses. The dose increase was associated with improvement in CML response in four of these five patients. In the five CML patients who had a cytogenetic response but did not have TKI dose escalation, the improvement in cytogenetic response was noticed while patients were on eltrombopag. Three CML patients had a switch in their TKI (Online Supplementary Table S1). All three of these patients had already some improvement in thrombocytopenia before switching their TKI, with the change indicated for other non-hematologic adverse events in one patient and the inefficacy of the TKI in the other two patients. None of the six patients with MF responded (i.e., none had a sustained increase in platelet count to ≥100x109/L); minor upward transient variations in platelet counts were seen in three patients (from 21x109/L to 28x109/L, 41x109/L to 55x109/L and from 65x109L to 75x109/L, respectively). Table 1. Baseline characteristics. Table 2. Eltrombopag dose distribution, mg per day (all patients). As of the date of this report, 20 patients were off study because of a lack of response (n=9), stem cell transplant (n=2), death (n=2), patient’s wish (n=1), adverse events (n=2), TKI discontinuation (n=1), loss to follow-up (n=1) and stable platelets (n=2). The two deaths on study were not related to treatment. One death was secondary to infectious complication in a patient with MF. The second death was secondary to hemorrhagic shock in a CML patient. This patient, treated with dasatinib, developed hepatosplenomegaly and ascites while on study but the etiology was not known. There was no evidence of portal vein thrombosis on CT abdomen/pelvis. Both eltrombopag and dasatinib were held and she had no platelets response. The platelet count was 10x109/L at the time of death due to severe gastrointestinal and genitourinary bleeding. Figure 1. Platelet change from baseline to response. *Each blue bar reflects change in platelet count in a chronic myeloid leukemia patient, while each green bar reflects change in platelet count in a myelofibrosis patient. Therapy was well tolerated in most patients, but two patients on ponatinib developed thrombotic events. Two months after eltrombopag discontinuation due to termination of the study, one patient with CML developed significant thrombocytosis (>1,000x109/L) with a white blood cell count of 9.5x109/L, 3% basophils, and 2% peripheral blast accompanied by headache and eye pain. Ophthalmoscopic examination was suggestive of bilateral plaques or thrombosis in the retinal vasculature but fluoroscopic evaluation did not reveal retinal vasculature blockage. Ponatinib was discontinued and thrombocytosis was managed with hydroxyurea. The aforementioned symptoms resolved. There was no cytogenetic response prior or after starting eltrombopag. Seven months after stopping eltrombopag, the patient had a persistent increase in blasts to 13% without a lack of hematologic response and she was then started on a clinical trial with an investigational TKI. Another CML patient developed non-occlusive deep venous thrombosis in the right upper extremity without thrombocytosis while on ponatinib 4 months after the study was terminated. One MF patient who had a history of coronary artery disease status post coronary artery bypass surgery developed myocardial infarction (MI) while on eltrombopag. This patent had then worsening increase in bone marrow fibrosis from grade 2 to grade 3 and was taken off study 40 days after MI. No further additional thrombotic/thromboembolic complications in CML and MF patients observed during or after the study (Online Supplementary Table S2). One patient (CML) discontinued therapy due to toxicity (elevation of liver function tests). Grade 3/4 toxicities irrespective of attribution listed in Table 4. One patient with MF had an increase in bone marrow fibrosis from grade 2 to grade 3. That patient had an increase in blast from 3% to 8% in the peripheral blood and an increase from 1% to 6% in the bone marrow while he was on study but with an improvement in hemoglobin. There was no change in the patient's disease other than this change in blast percentage. The patient was taken off study for lack of platelet response and later started on another clinical trial (PRM-151 + ruxolitinib). No progression of disease has been documented in any other patients. No clonal evolution was observed in patients with prolonged eltrombopag use. Discussion Thrombocytopenia is a common adverse event in patients with CML and MF who are treated with TKI and ruxolitinib, respectively.10,23 In most instances, thrombocytopenia is transient, occurs early during treatment initiation, and can be successfully managed with transient treatment interruptions and occasionally dose adjustments. However, in some patients thrombocytopenia can be persistent and more severe requiring frequent treatment interruptions and dose reductions, which might adversely influence treatment outcome.11 To that end, rIL-11 was successfully used in CML patients for the management of TKI associated thrombocytopenia.16 The main limitation of use of rIL-11 in the management of chemotherapy-induced thrombocytopenia in solid malignancies was the narrow therapeutic window with significant fluid retention and occasional arrhythmias. However, at lower doses used in CML, it was well tolerated24,25 with grade 1 or 2 peripheral edema observed in six patients (43%). Eltrombopag is a second generation oral thrombopoietin receptor agonist that has induced improvement of thrombocytopenia in patients with immune-mediate thrombocytopenia (ITP) or aplastic anemia. The EXTEND trial demonstrated that long-term use of eltrombopag was effective in maintaining for more than 6 months platelet counts of 50×109/L or more and reducing bleeding in most patients with ITP. Addition of eltrombopag to immunosuppressive treatment also markedly increased overall and complete hematologic response rates in treatment-naive severe aplastic anemia.26 Here we describe the use of eltrombopag in the management of TKI-related thrombocytopenia in CML and MF. Our results suggest clinical benefit in most patients with CML with a generally favorable safety profile, although two patents (both on ponartinib) had thrombotic events. In contrast, no response was observed in patients with MF. Theoretical concerns about the use of eltrombopag in this setting include increase in marrow blasts and possible transformation to advanced phases, thrombotic events including portal vein thrombosis, and increase in marrow fibrosis. We did not observe any instance of transformation in our series, in concordance with pre-clinical and clinical data showing no evidence of worsening leukemia.20,27 There was also no increase in marrow fibrosis in CML patients. Our series is small so the lack of such events should be considered as preliminary but reassuring. The most common adverse event was LFT elevation, but these were generally transient, reversible and manageable with dose adjustments. However, in one case it led to discontinuation of eltrombopag because of recurrent transaminitis. Two patients who received ponatinib (50%) had thrombotic events while on eltrombopag, this might raise the precaution of using ponatinib in conjunction with eltrombopag in CML patients. Despite the median disease duration of 2.2 years and multiple TKI use in CML patients before enrollment, eltrombopag demonstrated clinical efficacy with complete platelet response of 80% (12 of 15). This compares favorably to what was reported with rIL-11.16 More important, nine patients (60%) had improvement in cytogenetic responses, likely the result of a more sustained therapy with TKI. Notably, as doses of eltrombopag were increased, LFT elevations were noted in some patients. Conversely, eltrombopag dose interruptions or reductions due to such events or to platelets reaching >200x109/L, occasionally resulted in a drop-in platelet counts. Thus, close monitoring and dynamic management is required, at least during the initial stages of therapy to obtain the maximum effect while maintaining safety. The lack of efficacy among patients with MF could be in part secondary to advanced disease, or possible antagonism between the two medications. Thrombopoietin agonist are dependent on JAK-stat pathway which is inhibited by ruxolitinib.28 Table 3. Response to eltrombopag in chronic myeloid leukemia patients. Table 4. Treatment emergent adverse events. Our study has several limitations. It was a small study, and it did not accrue to the target sample size of 29 patients due to slow enrollment making the observation preliminary and requiring confirmation. We also do not have evidence or investigation of any immune mechanisms associated with thrombocytopenia, although we believe it is unlikely that these patients with CML had an immune mediated thrombocytopenia, and uncommon occurrence in this setting. In conclusion, our findings show that eltrombopag doses up to 300 mg may alleviate TKI-associated thrombocytopenia in some patients with CML. No similar benefit has been observed in patients with MF treated with ruxolitinib. Although generally safe, thrombotic events were noted that deserve further investigation, particularly when used in combination with ponatinib. Additional studies are warranted to confirm these observations. Supplementary Material Supplementary Appendix
DASATINIB
DrugsGivenReaction
CC BY-NC
33054123
20,176,531
2021-11-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Thrombocytopenia'.
The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis. Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKIs) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); median age 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12/15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients 5 could re-escalate the TKI dose and 9 improved their response. None of the 6 patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted. pmcIntroduction Tyrosine kinase inhibitors (TKI) are standard therapy for chronic myeloid leukemia (CML) and myelofibrosis (MF). Five TKI are currently approved for the treatment of CML in various stages, namely imatinib, nilotinib, dasatinib, bosutinib and ponatinib. Although these agents are generally well tolerated, some patients may develop adverse events, with myelosuppression being the most prominent.1-6 In most instances myelosuppression is grade 1 or 2 and requires no intervention. However, grade ≥3 thrombocytopenia (platelet ≤50x109/L) has been reported in 20% to 50% of patients. When this occurs, patients are usually managed with treatment interruption until platelets recovery (e.g., above 75x109/L) and dose reductions if thrombocytopenia recurs. Ruxolitinib is a JAK2 inhibitor used to manage splenomegaly and diseaseassociated symptoms in patients with MF.7 The dose limiting toxicity of ruxolitinib was thrombocytopenia8 and because of this the two pivotal phase III studies excluded patients with platelets ≤100x109/L. Still, thrombocytopenia was reported in 69% of patients, including 9% with grade ≥3.9 In patients with a platelet count of 50-100×109/L, grade ≥3 thrombocytopenia occurred in 56% of patients.10 Ruxolitinib-associated grade ≥3 thrombocytopenia is also typically managed with dose reductions or interruptions. Frequent dose interruptions and reductions might decrease TKI efficacy11,12 and may still not be sufficient to control thrombocytopenia. Patients who develop myelosuppression have a lower probability of achieving major or complete cytogenetic response (CCyR).11 For example, one study reported that patients treated with imatinib who developed grade ≥3 thrombocytopenia had a lower probability of CCyR compared to those who never developed thrombocytopenia (35% vs. 59%, P=0.02, respectively). Similarly, ruxolitinib efficacy is compromised with dose reductions and interruptions.12,13 In order to minimize dose reductions and interruptions, hematopoietic growth factors, filgrastim and erythropoietin stimulating agents (erythropoietin and darbepoetin) have been successfully used to manage neutropenia and anemia secondary to TKI in CML, respectively.14,15 Interleukin 11 (IL-11) was effective to manage thrombocytopenia associated with TKI in CML16 but use of this agent is associated with significant adverse events including fluid retention and cardiac arrhythmias. Eltrombopag is a non-peptide thrombopoietin receptor agonist that is effective and well tolerated among patients with immune thrombocytopenia, chronic hepatitis C-associated thrombocytopenia and severe aplastic anemia.17-19 It has also been safely used in acute myeloid leukemia without evidence of disease progression secondary to eltrombopag. 20 Here, we report the results from a pilot trial investigating the use of eltrombopag in the management of TKIor ruxolitinib-associated thrombocytopenia among patients with CML and MF. Methods Patients We conducted an open-label, non-randomized, phase II study of individualized dosing of eltrombopag. Eligible patients were aged 18 years or older with chronic phase CML receiving treatment with any Food and Drug Administration-approved TKI and experiencing grade ≥3 thrombocytopenia (platelets ≤50x109/L), or with MF receiving ruxolitinib and with platelets <100x109/L (since it is a dose-limiting toxicity and a label threshold for ruxolitinib), in either case after the first 3 months of therapy. Thrombocytopenia should have been either recurrent (i.e., be at least the second episode of thrombocytopenia) or have necessitated dose reductions of the TKI or ruxolitinib. All patients had signed an informed consent form approved by the Institutional Review Board, and the study was conducted in accordance with the Declaration of Helsinki. Study design Eltrombopag was commenced at 50 mg with dose escalation allowed every 2 weeks to 100 mg, 150 mg, 200 mg, and 300 mg (a higher dose than per label considering the thrombocytopenia refractoriness on these patients and the intent to continue TKI/ruxolitinib) according to platelet response. For patients of East Asian ancestry, eltrombopag was commenced at 25 mg daily with dose escalation allowed every 2 weeks. The following guideline was used to adjust dosing of eltrombopag: if the platelet count was >200x109/L at any time, the daily dose was reduced by 25 mg and re-assessed in 2 weeks; if >400x109/L, therapy was withheld and platelets assessed twice weekly until platelet count <150x109/L; therapy could then be resumed with the daily dose reduced by 25 mg. If the platelet count >400x109/L after 2 weeks was at the lowest dose, therapy was permanently discontinued. TKI doses were adjusted at the discretion of the treating physician per standard practice. Liver function tests (LFT) (alanine aminotransferase [ALT], aspartate aminotransferase [AST], and bilirubin) were done before the initiation of eltrombopag, every 2 weeks during the dose adjustment phase and following the monthly establishment of a stable dose. When LFT abnormalities were identified, LFT were performed weekly until the abnormalities resolved or stabilized; if ALT/AST levels were ≥ three-times the upper limit of normal (ULN): therapy was withheld, we then repeated abnormal liver function tests within 3-5 days; if confirmed abnormal, we monitored LFT weekly until resolved, stabilized, or returned to baseline. If ALT/AST levels ≥ three-times the ULN and were progressive, persistent (≥4 weeks), accompanied by increased direct bilirubin, or accompanied by clinical signs of liver injury or evidence of hepatic decompensation, eltrombopag was permanently discontinued. Patients who experienced other clinically significant grade 3 or greater toxicity possibly related to eltrombopag, had eltrombopag interruption until toxicity resolved to grade 1 or less. Treatment then was resumed at the immediate lower dose level. Failure to achieve a platelet count ≥50x109/L or ≥100x109/L in CML and MF patients, respectively after 8 weeks of eltrombopag was considered as lack of response. Statistical analysis Simon’s optimal two-stage design (Simon, 1989) was used to test the null hypothesis that the proportion of subjects with complete response is ≤0.10 versus the alternative that it is ≥0.30 (i.e., Po≤0.10 vs. Pa≥0.30) at alpha=0.05 with 80% power. The design resulted in an expected sample size of 15 and a probability of early termination of 0.736. The study was designed to study eltrombopag in ten patients in the first stage; the trial would be terminated if one or fewer achieved complete platelet response. Otherwise, the trial would go to the second stage, and 29 patients would be studied. If the total number of patients with complete platelet response were less than or equal to five, the drug would be deemed ineffective. The MF group was an exploratory group of ten patients to study the safety and activity of eltrombopag in patients with MF treated with ruxolitinib. We considered the activity promising if three or more patients out of ten achieved complete platelet response. For safety monitoring in the cohort with MF, accrual would stop if, at any time, four of ten patients encounter grade 3 or more nonhematological toxicity or progression to acute leukemia. As an additional safety procedure, we observed the first three MF patients on trial for at least 3 months before other patients were accrued. Response definitions Complete platelet response was defined as platelet count ≥50x109/L for CML, and ≥100x109/L for MF that was sustained for ≥3 months while continuing TKI or ruxolitinib therapy or with sustained (≥3 months) re-escalation of TKI dose without recurrence of thrombocytopenia. Criteria for CML and MF response were previously defined.21,22 The target response was a complete response in at least 30% of patients. Results Twenty-one patients were enrolled: 15 with CML and six with MF. Their median age was 60 years (range, 31-97) and their clinical characteristics are shown in Table 1. Median duration of disease was 2.2 years (range, 0.5-29 years) for patients with CML and 2 years (range, 0.3-3.6 years) for patients with MF. At the time of enrollment, patients with CML were receiving the following TKI: dasatinib (n=5), ponatinib (n=4), nilotinib (n=3), bosutinib (n=2), and imatinib (n=1), 27% were receiving their first TKI, 27% the second TKI, 27% the third, and 19% the fourth or later TKI. The median platelet count was 44x109/L (range, 3-49x109/L) in patients with CML and 62x109/L (range, 21-75x109/L) in those with MF. Cytogenetic response for patients with CML at baseline were partial in three, minor in six, and none in six. Prior therapies in MF patients were an investigational JAK2 inhibitor, and interferon a-2 in one patient each. The median dose of ruxolitinib was 10 mg (range, 10-30 mg) (Table 1). Eltrombopag dose distribution is summarized in Table 2. After a median duration of treatment of 18 months (range, 5-77 months), 12 of the 15 (80%) patients with CML achieved a complete platelet response with doses of eltrombopag of 50–300 mg per day. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). The median time to best response was 6 months (range, 2.1-13 months). Ten patients had sustained platelet recovery after stopping eltrombopag. The median duration for sustained platelet response was 45 months (range, 3-69 months). The three patients who did not achieve a complete platelet response had only minor changes in platelet count while they were taking eltrombopag (from 3x109/L to 8x109/L, 19x109/L to 45x109/L, and from 42x109/L to 46x109/L, respectively). Two patients (one each of CML and MF) had improvement in hemoglobin of over 2 g/dL from baseline (from 8.2 g/dL to 10.6 g/dL, and from 9.4 g/dL to 11.4 g/dL, respectively), Hemoglobin improvement was sustained over 21.5 and 2 months respectively while patients were taking eltrombopag. Hemoglobin levels declined after interruption of eltrombopag. One patient with CML had an absolute neutrophil count recovery to >1x109/L (baseline neutrophils 0.71x109/L). Absolute neutrophil count improvement was sustained for >6 months while on eltrombopag. Absolute neutrophil count then declined after interruption of eltrombopag. The TKI doses and duration for patients with CML post enrollment are summarized in Table 1. Nine patients with CML experienced an improvement in the cytogenetic response during the observation period (all of them had sustained platelet recovery after stopping eltrombopag); one from none to complete, two from minor to complete, four from minor to partial, and two from partial to complete (Table 3). In five patients with CML the TKI dose was increased and maintained while continuing eltrombopag. Dasatinib daily dose was increased from 50 mg to 100 mg in three patients, nilotinib dose was increased in one patient form 150 mg twice daily to 200 mg twice daily, and one patient had an increase in ponatinib dose from 15 mg every other day to 15 mg daily. There were no TKI dose-limiting toxicities in patients who increased their TKI doses. The dose increase was associated with improvement in CML response in four of these five patients. In the five CML patients who had a cytogenetic response but did not have TKI dose escalation, the improvement in cytogenetic response was noticed while patients were on eltrombopag. Three CML patients had a switch in their TKI (Online Supplementary Table S1). All three of these patients had already some improvement in thrombocytopenia before switching their TKI, with the change indicated for other non-hematologic adverse events in one patient and the inefficacy of the TKI in the other two patients. None of the six patients with MF responded (i.e., none had a sustained increase in platelet count to ≥100x109/L); minor upward transient variations in platelet counts were seen in three patients (from 21x109/L to 28x109/L, 41x109/L to 55x109/L and from 65x109L to 75x109/L, respectively). Table 1. Baseline characteristics. Table 2. Eltrombopag dose distribution, mg per day (all patients). As of the date of this report, 20 patients were off study because of a lack of response (n=9), stem cell transplant (n=2), death (n=2), patient’s wish (n=1), adverse events (n=2), TKI discontinuation (n=1), loss to follow-up (n=1) and stable platelets (n=2). The two deaths on study were not related to treatment. One death was secondary to infectious complication in a patient with MF. The second death was secondary to hemorrhagic shock in a CML patient. This patient, treated with dasatinib, developed hepatosplenomegaly and ascites while on study but the etiology was not known. There was no evidence of portal vein thrombosis on CT abdomen/pelvis. Both eltrombopag and dasatinib were held and she had no platelets response. The platelet count was 10x109/L at the time of death due to severe gastrointestinal and genitourinary bleeding. Figure 1. Platelet change from baseline to response. *Each blue bar reflects change in platelet count in a chronic myeloid leukemia patient, while each green bar reflects change in platelet count in a myelofibrosis patient. Therapy was well tolerated in most patients, but two patients on ponatinib developed thrombotic events. Two months after eltrombopag discontinuation due to termination of the study, one patient with CML developed significant thrombocytosis (>1,000x109/L) with a white blood cell count of 9.5x109/L, 3% basophils, and 2% peripheral blast accompanied by headache and eye pain. Ophthalmoscopic examination was suggestive of bilateral plaques or thrombosis in the retinal vasculature but fluoroscopic evaluation did not reveal retinal vasculature blockage. Ponatinib was discontinued and thrombocytosis was managed with hydroxyurea. The aforementioned symptoms resolved. There was no cytogenetic response prior or after starting eltrombopag. Seven months after stopping eltrombopag, the patient had a persistent increase in blasts to 13% without a lack of hematologic response and she was then started on a clinical trial with an investigational TKI. Another CML patient developed non-occlusive deep venous thrombosis in the right upper extremity without thrombocytosis while on ponatinib 4 months after the study was terminated. One MF patient who had a history of coronary artery disease status post coronary artery bypass surgery developed myocardial infarction (MI) while on eltrombopag. This patent had then worsening increase in bone marrow fibrosis from grade 2 to grade 3 and was taken off study 40 days after MI. No further additional thrombotic/thromboembolic complications in CML and MF patients observed during or after the study (Online Supplementary Table S2). One patient (CML) discontinued therapy due to toxicity (elevation of liver function tests). Grade 3/4 toxicities irrespective of attribution listed in Table 4. One patient with MF had an increase in bone marrow fibrosis from grade 2 to grade 3. That patient had an increase in blast from 3% to 8% in the peripheral blood and an increase from 1% to 6% in the bone marrow while he was on study but with an improvement in hemoglobin. There was no change in the patient's disease other than this change in blast percentage. The patient was taken off study for lack of platelet response and later started on another clinical trial (PRM-151 + ruxolitinib). No progression of disease has been documented in any other patients. No clonal evolution was observed in patients with prolonged eltrombopag use. Discussion Thrombocytopenia is a common adverse event in patients with CML and MF who are treated with TKI and ruxolitinib, respectively.10,23 In most instances, thrombocytopenia is transient, occurs early during treatment initiation, and can be successfully managed with transient treatment interruptions and occasionally dose adjustments. However, in some patients thrombocytopenia can be persistent and more severe requiring frequent treatment interruptions and dose reductions, which might adversely influence treatment outcome.11 To that end, rIL-11 was successfully used in CML patients for the management of TKI associated thrombocytopenia.16 The main limitation of use of rIL-11 in the management of chemotherapy-induced thrombocytopenia in solid malignancies was the narrow therapeutic window with significant fluid retention and occasional arrhythmias. However, at lower doses used in CML, it was well tolerated24,25 with grade 1 or 2 peripheral edema observed in six patients (43%). Eltrombopag is a second generation oral thrombopoietin receptor agonist that has induced improvement of thrombocytopenia in patients with immune-mediate thrombocytopenia (ITP) or aplastic anemia. The EXTEND trial demonstrated that long-term use of eltrombopag was effective in maintaining for more than 6 months platelet counts of 50×109/L or more and reducing bleeding in most patients with ITP. Addition of eltrombopag to immunosuppressive treatment also markedly increased overall and complete hematologic response rates in treatment-naive severe aplastic anemia.26 Here we describe the use of eltrombopag in the management of TKI-related thrombocytopenia in CML and MF. Our results suggest clinical benefit in most patients with CML with a generally favorable safety profile, although two patents (both on ponartinib) had thrombotic events. In contrast, no response was observed in patients with MF. Theoretical concerns about the use of eltrombopag in this setting include increase in marrow blasts and possible transformation to advanced phases, thrombotic events including portal vein thrombosis, and increase in marrow fibrosis. We did not observe any instance of transformation in our series, in concordance with pre-clinical and clinical data showing no evidence of worsening leukemia.20,27 There was also no increase in marrow fibrosis in CML patients. Our series is small so the lack of such events should be considered as preliminary but reassuring. The most common adverse event was LFT elevation, but these were generally transient, reversible and manageable with dose adjustments. However, in one case it led to discontinuation of eltrombopag because of recurrent transaminitis. Two patients who received ponatinib (50%) had thrombotic events while on eltrombopag, this might raise the precaution of using ponatinib in conjunction with eltrombopag in CML patients. Despite the median disease duration of 2.2 years and multiple TKI use in CML patients before enrollment, eltrombopag demonstrated clinical efficacy with complete platelet response of 80% (12 of 15). This compares favorably to what was reported with rIL-11.16 More important, nine patients (60%) had improvement in cytogenetic responses, likely the result of a more sustained therapy with TKI. Notably, as doses of eltrombopag were increased, LFT elevations were noted in some patients. Conversely, eltrombopag dose interruptions or reductions due to such events or to platelets reaching >200x109/L, occasionally resulted in a drop-in platelet counts. Thus, close monitoring and dynamic management is required, at least during the initial stages of therapy to obtain the maximum effect while maintaining safety. The lack of efficacy among patients with MF could be in part secondary to advanced disease, or possible antagonism between the two medications. Thrombopoietin agonist are dependent on JAK-stat pathway which is inhibited by ruxolitinib.28 Table 3. Response to eltrombopag in chronic myeloid leukemia patients. Table 4. Treatment emergent adverse events. Our study has several limitations. It was a small study, and it did not accrue to the target sample size of 29 patients due to slow enrollment making the observation preliminary and requiring confirmation. We also do not have evidence or investigation of any immune mechanisms associated with thrombocytopenia, although we believe it is unlikely that these patients with CML had an immune mediated thrombocytopenia, and uncommon occurrence in this setting. In conclusion, our findings show that eltrombopag doses up to 300 mg may alleviate TKI-associated thrombocytopenia in some patients with CML. No similar benefit has been observed in patients with MF treated with ruxolitinib. Although generally safe, thrombotic events were noted that deserve further investigation, particularly when used in combination with ponatinib. Additional studies are warranted to confirm these observations. Supplementary Material Supplementary Appendix
DASATINIB
DrugsGivenReaction
CC BY-NC
33054123
20,176,531
2021-11-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Urogenital haemorrhage'.
The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis. Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKIs) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); median age 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12/15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients 5 could re-escalate the TKI dose and 9 improved their response. None of the 6 patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted. pmcIntroduction Tyrosine kinase inhibitors (TKI) are standard therapy for chronic myeloid leukemia (CML) and myelofibrosis (MF). Five TKI are currently approved for the treatment of CML in various stages, namely imatinib, nilotinib, dasatinib, bosutinib and ponatinib. Although these agents are generally well tolerated, some patients may develop adverse events, with myelosuppression being the most prominent.1-6 In most instances myelosuppression is grade 1 or 2 and requires no intervention. However, grade ≥3 thrombocytopenia (platelet ≤50x109/L) has been reported in 20% to 50% of patients. When this occurs, patients are usually managed with treatment interruption until platelets recovery (e.g., above 75x109/L) and dose reductions if thrombocytopenia recurs. Ruxolitinib is a JAK2 inhibitor used to manage splenomegaly and diseaseassociated symptoms in patients with MF.7 The dose limiting toxicity of ruxolitinib was thrombocytopenia8 and because of this the two pivotal phase III studies excluded patients with platelets ≤100x109/L. Still, thrombocytopenia was reported in 69% of patients, including 9% with grade ≥3.9 In patients with a platelet count of 50-100×109/L, grade ≥3 thrombocytopenia occurred in 56% of patients.10 Ruxolitinib-associated grade ≥3 thrombocytopenia is also typically managed with dose reductions or interruptions. Frequent dose interruptions and reductions might decrease TKI efficacy11,12 and may still not be sufficient to control thrombocytopenia. Patients who develop myelosuppression have a lower probability of achieving major or complete cytogenetic response (CCyR).11 For example, one study reported that patients treated with imatinib who developed grade ≥3 thrombocytopenia had a lower probability of CCyR compared to those who never developed thrombocytopenia (35% vs. 59%, P=0.02, respectively). Similarly, ruxolitinib efficacy is compromised with dose reductions and interruptions.12,13 In order to minimize dose reductions and interruptions, hematopoietic growth factors, filgrastim and erythropoietin stimulating agents (erythropoietin and darbepoetin) have been successfully used to manage neutropenia and anemia secondary to TKI in CML, respectively.14,15 Interleukin 11 (IL-11) was effective to manage thrombocytopenia associated with TKI in CML16 but use of this agent is associated with significant adverse events including fluid retention and cardiac arrhythmias. Eltrombopag is a non-peptide thrombopoietin receptor agonist that is effective and well tolerated among patients with immune thrombocytopenia, chronic hepatitis C-associated thrombocytopenia and severe aplastic anemia.17-19 It has also been safely used in acute myeloid leukemia without evidence of disease progression secondary to eltrombopag. 20 Here, we report the results from a pilot trial investigating the use of eltrombopag in the management of TKIor ruxolitinib-associated thrombocytopenia among patients with CML and MF. Methods Patients We conducted an open-label, non-randomized, phase II study of individualized dosing of eltrombopag. Eligible patients were aged 18 years or older with chronic phase CML receiving treatment with any Food and Drug Administration-approved TKI and experiencing grade ≥3 thrombocytopenia (platelets ≤50x109/L), or with MF receiving ruxolitinib and with platelets <100x109/L (since it is a dose-limiting toxicity and a label threshold for ruxolitinib), in either case after the first 3 months of therapy. Thrombocytopenia should have been either recurrent (i.e., be at least the second episode of thrombocytopenia) or have necessitated dose reductions of the TKI or ruxolitinib. All patients had signed an informed consent form approved by the Institutional Review Board, and the study was conducted in accordance with the Declaration of Helsinki. Study design Eltrombopag was commenced at 50 mg with dose escalation allowed every 2 weeks to 100 mg, 150 mg, 200 mg, and 300 mg (a higher dose than per label considering the thrombocytopenia refractoriness on these patients and the intent to continue TKI/ruxolitinib) according to platelet response. For patients of East Asian ancestry, eltrombopag was commenced at 25 mg daily with dose escalation allowed every 2 weeks. The following guideline was used to adjust dosing of eltrombopag: if the platelet count was >200x109/L at any time, the daily dose was reduced by 25 mg and re-assessed in 2 weeks; if >400x109/L, therapy was withheld and platelets assessed twice weekly until platelet count <150x109/L; therapy could then be resumed with the daily dose reduced by 25 mg. If the platelet count >400x109/L after 2 weeks was at the lowest dose, therapy was permanently discontinued. TKI doses were adjusted at the discretion of the treating physician per standard practice. Liver function tests (LFT) (alanine aminotransferase [ALT], aspartate aminotransferase [AST], and bilirubin) were done before the initiation of eltrombopag, every 2 weeks during the dose adjustment phase and following the monthly establishment of a stable dose. When LFT abnormalities were identified, LFT were performed weekly until the abnormalities resolved or stabilized; if ALT/AST levels were ≥ three-times the upper limit of normal (ULN): therapy was withheld, we then repeated abnormal liver function tests within 3-5 days; if confirmed abnormal, we monitored LFT weekly until resolved, stabilized, or returned to baseline. If ALT/AST levels ≥ three-times the ULN and were progressive, persistent (≥4 weeks), accompanied by increased direct bilirubin, or accompanied by clinical signs of liver injury or evidence of hepatic decompensation, eltrombopag was permanently discontinued. Patients who experienced other clinically significant grade 3 or greater toxicity possibly related to eltrombopag, had eltrombopag interruption until toxicity resolved to grade 1 or less. Treatment then was resumed at the immediate lower dose level. Failure to achieve a platelet count ≥50x109/L or ≥100x109/L in CML and MF patients, respectively after 8 weeks of eltrombopag was considered as lack of response. Statistical analysis Simon’s optimal two-stage design (Simon, 1989) was used to test the null hypothesis that the proportion of subjects with complete response is ≤0.10 versus the alternative that it is ≥0.30 (i.e., Po≤0.10 vs. Pa≥0.30) at alpha=0.05 with 80% power. The design resulted in an expected sample size of 15 and a probability of early termination of 0.736. The study was designed to study eltrombopag in ten patients in the first stage; the trial would be terminated if one or fewer achieved complete platelet response. Otherwise, the trial would go to the second stage, and 29 patients would be studied. If the total number of patients with complete platelet response were less than or equal to five, the drug would be deemed ineffective. The MF group was an exploratory group of ten patients to study the safety and activity of eltrombopag in patients with MF treated with ruxolitinib. We considered the activity promising if three or more patients out of ten achieved complete platelet response. For safety monitoring in the cohort with MF, accrual would stop if, at any time, four of ten patients encounter grade 3 or more nonhematological toxicity or progression to acute leukemia. As an additional safety procedure, we observed the first three MF patients on trial for at least 3 months before other patients were accrued. Response definitions Complete platelet response was defined as platelet count ≥50x109/L for CML, and ≥100x109/L for MF that was sustained for ≥3 months while continuing TKI or ruxolitinib therapy or with sustained (≥3 months) re-escalation of TKI dose without recurrence of thrombocytopenia. Criteria for CML and MF response were previously defined.21,22 The target response was a complete response in at least 30% of patients. Results Twenty-one patients were enrolled: 15 with CML and six with MF. Their median age was 60 years (range, 31-97) and their clinical characteristics are shown in Table 1. Median duration of disease was 2.2 years (range, 0.5-29 years) for patients with CML and 2 years (range, 0.3-3.6 years) for patients with MF. At the time of enrollment, patients with CML were receiving the following TKI: dasatinib (n=5), ponatinib (n=4), nilotinib (n=3), bosutinib (n=2), and imatinib (n=1), 27% were receiving their first TKI, 27% the second TKI, 27% the third, and 19% the fourth or later TKI. The median platelet count was 44x109/L (range, 3-49x109/L) in patients with CML and 62x109/L (range, 21-75x109/L) in those with MF. Cytogenetic response for patients with CML at baseline were partial in three, minor in six, and none in six. Prior therapies in MF patients were an investigational JAK2 inhibitor, and interferon a-2 in one patient each. The median dose of ruxolitinib was 10 mg (range, 10-30 mg) (Table 1). Eltrombopag dose distribution is summarized in Table 2. After a median duration of treatment of 18 months (range, 5-77 months), 12 of the 15 (80%) patients with CML achieved a complete platelet response with doses of eltrombopag of 50–300 mg per day. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). The median time to best response was 6 months (range, 2.1-13 months). Ten patients had sustained platelet recovery after stopping eltrombopag. The median duration for sustained platelet response was 45 months (range, 3-69 months). The three patients who did not achieve a complete platelet response had only minor changes in platelet count while they were taking eltrombopag (from 3x109/L to 8x109/L, 19x109/L to 45x109/L, and from 42x109/L to 46x109/L, respectively). Two patients (one each of CML and MF) had improvement in hemoglobin of over 2 g/dL from baseline (from 8.2 g/dL to 10.6 g/dL, and from 9.4 g/dL to 11.4 g/dL, respectively), Hemoglobin improvement was sustained over 21.5 and 2 months respectively while patients were taking eltrombopag. Hemoglobin levels declined after interruption of eltrombopag. One patient with CML had an absolute neutrophil count recovery to >1x109/L (baseline neutrophils 0.71x109/L). Absolute neutrophil count improvement was sustained for >6 months while on eltrombopag. Absolute neutrophil count then declined after interruption of eltrombopag. The TKI doses and duration for patients with CML post enrollment are summarized in Table 1. Nine patients with CML experienced an improvement in the cytogenetic response during the observation period (all of them had sustained platelet recovery after stopping eltrombopag); one from none to complete, two from minor to complete, four from minor to partial, and two from partial to complete (Table 3). In five patients with CML the TKI dose was increased and maintained while continuing eltrombopag. Dasatinib daily dose was increased from 50 mg to 100 mg in three patients, nilotinib dose was increased in one patient form 150 mg twice daily to 200 mg twice daily, and one patient had an increase in ponatinib dose from 15 mg every other day to 15 mg daily. There were no TKI dose-limiting toxicities in patients who increased their TKI doses. The dose increase was associated with improvement in CML response in four of these five patients. In the five CML patients who had a cytogenetic response but did not have TKI dose escalation, the improvement in cytogenetic response was noticed while patients were on eltrombopag. Three CML patients had a switch in their TKI (Online Supplementary Table S1). All three of these patients had already some improvement in thrombocytopenia before switching their TKI, with the change indicated for other non-hematologic adverse events in one patient and the inefficacy of the TKI in the other two patients. None of the six patients with MF responded (i.e., none had a sustained increase in platelet count to ≥100x109/L); minor upward transient variations in platelet counts were seen in three patients (from 21x109/L to 28x109/L, 41x109/L to 55x109/L and from 65x109L to 75x109/L, respectively). Table 1. Baseline characteristics. Table 2. Eltrombopag dose distribution, mg per day (all patients). As of the date of this report, 20 patients were off study because of a lack of response (n=9), stem cell transplant (n=2), death (n=2), patient’s wish (n=1), adverse events (n=2), TKI discontinuation (n=1), loss to follow-up (n=1) and stable platelets (n=2). The two deaths on study were not related to treatment. One death was secondary to infectious complication in a patient with MF. The second death was secondary to hemorrhagic shock in a CML patient. This patient, treated with dasatinib, developed hepatosplenomegaly and ascites while on study but the etiology was not known. There was no evidence of portal vein thrombosis on CT abdomen/pelvis. Both eltrombopag and dasatinib were held and she had no platelets response. The platelet count was 10x109/L at the time of death due to severe gastrointestinal and genitourinary bleeding. Figure 1. Platelet change from baseline to response. *Each blue bar reflects change in platelet count in a chronic myeloid leukemia patient, while each green bar reflects change in platelet count in a myelofibrosis patient. Therapy was well tolerated in most patients, but two patients on ponatinib developed thrombotic events. Two months after eltrombopag discontinuation due to termination of the study, one patient with CML developed significant thrombocytosis (>1,000x109/L) with a white blood cell count of 9.5x109/L, 3% basophils, and 2% peripheral blast accompanied by headache and eye pain. Ophthalmoscopic examination was suggestive of bilateral plaques or thrombosis in the retinal vasculature but fluoroscopic evaluation did not reveal retinal vasculature blockage. Ponatinib was discontinued and thrombocytosis was managed with hydroxyurea. The aforementioned symptoms resolved. There was no cytogenetic response prior or after starting eltrombopag. Seven months after stopping eltrombopag, the patient had a persistent increase in blasts to 13% without a lack of hematologic response and she was then started on a clinical trial with an investigational TKI. Another CML patient developed non-occlusive deep venous thrombosis in the right upper extremity without thrombocytosis while on ponatinib 4 months after the study was terminated. One MF patient who had a history of coronary artery disease status post coronary artery bypass surgery developed myocardial infarction (MI) while on eltrombopag. This patent had then worsening increase in bone marrow fibrosis from grade 2 to grade 3 and was taken off study 40 days after MI. No further additional thrombotic/thromboembolic complications in CML and MF patients observed during or after the study (Online Supplementary Table S2). One patient (CML) discontinued therapy due to toxicity (elevation of liver function tests). Grade 3/4 toxicities irrespective of attribution listed in Table 4. One patient with MF had an increase in bone marrow fibrosis from grade 2 to grade 3. That patient had an increase in blast from 3% to 8% in the peripheral blood and an increase from 1% to 6% in the bone marrow while he was on study but with an improvement in hemoglobin. There was no change in the patient's disease other than this change in blast percentage. The patient was taken off study for lack of platelet response and later started on another clinical trial (PRM-151 + ruxolitinib). No progression of disease has been documented in any other patients. No clonal evolution was observed in patients with prolonged eltrombopag use. Discussion Thrombocytopenia is a common adverse event in patients with CML and MF who are treated with TKI and ruxolitinib, respectively.10,23 In most instances, thrombocytopenia is transient, occurs early during treatment initiation, and can be successfully managed with transient treatment interruptions and occasionally dose adjustments. However, in some patients thrombocytopenia can be persistent and more severe requiring frequent treatment interruptions and dose reductions, which might adversely influence treatment outcome.11 To that end, rIL-11 was successfully used in CML patients for the management of TKI associated thrombocytopenia.16 The main limitation of use of rIL-11 in the management of chemotherapy-induced thrombocytopenia in solid malignancies was the narrow therapeutic window with significant fluid retention and occasional arrhythmias. However, at lower doses used in CML, it was well tolerated24,25 with grade 1 or 2 peripheral edema observed in six patients (43%). Eltrombopag is a second generation oral thrombopoietin receptor agonist that has induced improvement of thrombocytopenia in patients with immune-mediate thrombocytopenia (ITP) or aplastic anemia. The EXTEND trial demonstrated that long-term use of eltrombopag was effective in maintaining for more than 6 months platelet counts of 50×109/L or more and reducing bleeding in most patients with ITP. Addition of eltrombopag to immunosuppressive treatment also markedly increased overall and complete hematologic response rates in treatment-naive severe aplastic anemia.26 Here we describe the use of eltrombopag in the management of TKI-related thrombocytopenia in CML and MF. Our results suggest clinical benefit in most patients with CML with a generally favorable safety profile, although two patents (both on ponartinib) had thrombotic events. In contrast, no response was observed in patients with MF. Theoretical concerns about the use of eltrombopag in this setting include increase in marrow blasts and possible transformation to advanced phases, thrombotic events including portal vein thrombosis, and increase in marrow fibrosis. We did not observe any instance of transformation in our series, in concordance with pre-clinical and clinical data showing no evidence of worsening leukemia.20,27 There was also no increase in marrow fibrosis in CML patients. Our series is small so the lack of such events should be considered as preliminary but reassuring. The most common adverse event was LFT elevation, but these were generally transient, reversible and manageable with dose adjustments. However, in one case it led to discontinuation of eltrombopag because of recurrent transaminitis. Two patients who received ponatinib (50%) had thrombotic events while on eltrombopag, this might raise the precaution of using ponatinib in conjunction with eltrombopag in CML patients. Despite the median disease duration of 2.2 years and multiple TKI use in CML patients before enrollment, eltrombopag demonstrated clinical efficacy with complete platelet response of 80% (12 of 15). This compares favorably to what was reported with rIL-11.16 More important, nine patients (60%) had improvement in cytogenetic responses, likely the result of a more sustained therapy with TKI. Notably, as doses of eltrombopag were increased, LFT elevations were noted in some patients. Conversely, eltrombopag dose interruptions or reductions due to such events or to platelets reaching >200x109/L, occasionally resulted in a drop-in platelet counts. Thus, close monitoring and dynamic management is required, at least during the initial stages of therapy to obtain the maximum effect while maintaining safety. The lack of efficacy among patients with MF could be in part secondary to advanced disease, or possible antagonism between the two medications. Thrombopoietin agonist are dependent on JAK-stat pathway which is inhibited by ruxolitinib.28 Table 3. Response to eltrombopag in chronic myeloid leukemia patients. Table 4. Treatment emergent adverse events. Our study has several limitations. It was a small study, and it did not accrue to the target sample size of 29 patients due to slow enrollment making the observation preliminary and requiring confirmation. We also do not have evidence or investigation of any immune mechanisms associated with thrombocytopenia, although we believe it is unlikely that these patients with CML had an immune mediated thrombocytopenia, and uncommon occurrence in this setting. In conclusion, our findings show that eltrombopag doses up to 300 mg may alleviate TKI-associated thrombocytopenia in some patients with CML. No similar benefit has been observed in patients with MF treated with ruxolitinib. Although generally safe, thrombotic events were noted that deserve further investigation, particularly when used in combination with ponatinib. Additional studies are warranted to confirm these observations. Supplementary Material Supplementary Appendix
DASATINIB
DrugsGivenReaction
CC BY-NC
33054123
20,176,531
2021-11-01
What was the outcome of reaction 'Gastrointestinal haemorrhage'?
The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis. Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKIs) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); median age 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12/15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients 5 could re-escalate the TKI dose and 9 improved their response. None of the 6 patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted. pmcIntroduction Tyrosine kinase inhibitors (TKI) are standard therapy for chronic myeloid leukemia (CML) and myelofibrosis (MF). Five TKI are currently approved for the treatment of CML in various stages, namely imatinib, nilotinib, dasatinib, bosutinib and ponatinib. Although these agents are generally well tolerated, some patients may develop adverse events, with myelosuppression being the most prominent.1-6 In most instances myelosuppression is grade 1 or 2 and requires no intervention. However, grade ≥3 thrombocytopenia (platelet ≤50x109/L) has been reported in 20% to 50% of patients. When this occurs, patients are usually managed with treatment interruption until platelets recovery (e.g., above 75x109/L) and dose reductions if thrombocytopenia recurs. Ruxolitinib is a JAK2 inhibitor used to manage splenomegaly and diseaseassociated symptoms in patients with MF.7 The dose limiting toxicity of ruxolitinib was thrombocytopenia8 and because of this the two pivotal phase III studies excluded patients with platelets ≤100x109/L. Still, thrombocytopenia was reported in 69% of patients, including 9% with grade ≥3.9 In patients with a platelet count of 50-100×109/L, grade ≥3 thrombocytopenia occurred in 56% of patients.10 Ruxolitinib-associated grade ≥3 thrombocytopenia is also typically managed with dose reductions or interruptions. Frequent dose interruptions and reductions might decrease TKI efficacy11,12 and may still not be sufficient to control thrombocytopenia. Patients who develop myelosuppression have a lower probability of achieving major or complete cytogenetic response (CCyR).11 For example, one study reported that patients treated with imatinib who developed grade ≥3 thrombocytopenia had a lower probability of CCyR compared to those who never developed thrombocytopenia (35% vs. 59%, P=0.02, respectively). Similarly, ruxolitinib efficacy is compromised with dose reductions and interruptions.12,13 In order to minimize dose reductions and interruptions, hematopoietic growth factors, filgrastim and erythropoietin stimulating agents (erythropoietin and darbepoetin) have been successfully used to manage neutropenia and anemia secondary to TKI in CML, respectively.14,15 Interleukin 11 (IL-11) was effective to manage thrombocytopenia associated with TKI in CML16 but use of this agent is associated with significant adverse events including fluid retention and cardiac arrhythmias. Eltrombopag is a non-peptide thrombopoietin receptor agonist that is effective and well tolerated among patients with immune thrombocytopenia, chronic hepatitis C-associated thrombocytopenia and severe aplastic anemia.17-19 It has also been safely used in acute myeloid leukemia without evidence of disease progression secondary to eltrombopag. 20 Here, we report the results from a pilot trial investigating the use of eltrombopag in the management of TKIor ruxolitinib-associated thrombocytopenia among patients with CML and MF. Methods Patients We conducted an open-label, non-randomized, phase II study of individualized dosing of eltrombopag. Eligible patients were aged 18 years or older with chronic phase CML receiving treatment with any Food and Drug Administration-approved TKI and experiencing grade ≥3 thrombocytopenia (platelets ≤50x109/L), or with MF receiving ruxolitinib and with platelets <100x109/L (since it is a dose-limiting toxicity and a label threshold for ruxolitinib), in either case after the first 3 months of therapy. Thrombocytopenia should have been either recurrent (i.e., be at least the second episode of thrombocytopenia) or have necessitated dose reductions of the TKI or ruxolitinib. All patients had signed an informed consent form approved by the Institutional Review Board, and the study was conducted in accordance with the Declaration of Helsinki. Study design Eltrombopag was commenced at 50 mg with dose escalation allowed every 2 weeks to 100 mg, 150 mg, 200 mg, and 300 mg (a higher dose than per label considering the thrombocytopenia refractoriness on these patients and the intent to continue TKI/ruxolitinib) according to platelet response. For patients of East Asian ancestry, eltrombopag was commenced at 25 mg daily with dose escalation allowed every 2 weeks. The following guideline was used to adjust dosing of eltrombopag: if the platelet count was >200x109/L at any time, the daily dose was reduced by 25 mg and re-assessed in 2 weeks; if >400x109/L, therapy was withheld and platelets assessed twice weekly until platelet count <150x109/L; therapy could then be resumed with the daily dose reduced by 25 mg. If the platelet count >400x109/L after 2 weeks was at the lowest dose, therapy was permanently discontinued. TKI doses were adjusted at the discretion of the treating physician per standard practice. Liver function tests (LFT) (alanine aminotransferase [ALT], aspartate aminotransferase [AST], and bilirubin) were done before the initiation of eltrombopag, every 2 weeks during the dose adjustment phase and following the monthly establishment of a stable dose. When LFT abnormalities were identified, LFT were performed weekly until the abnormalities resolved or stabilized; if ALT/AST levels were ≥ three-times the upper limit of normal (ULN): therapy was withheld, we then repeated abnormal liver function tests within 3-5 days; if confirmed abnormal, we monitored LFT weekly until resolved, stabilized, or returned to baseline. If ALT/AST levels ≥ three-times the ULN and were progressive, persistent (≥4 weeks), accompanied by increased direct bilirubin, or accompanied by clinical signs of liver injury or evidence of hepatic decompensation, eltrombopag was permanently discontinued. Patients who experienced other clinically significant grade 3 or greater toxicity possibly related to eltrombopag, had eltrombopag interruption until toxicity resolved to grade 1 or less. Treatment then was resumed at the immediate lower dose level. Failure to achieve a platelet count ≥50x109/L or ≥100x109/L in CML and MF patients, respectively after 8 weeks of eltrombopag was considered as lack of response. Statistical analysis Simon’s optimal two-stage design (Simon, 1989) was used to test the null hypothesis that the proportion of subjects with complete response is ≤0.10 versus the alternative that it is ≥0.30 (i.e., Po≤0.10 vs. Pa≥0.30) at alpha=0.05 with 80% power. The design resulted in an expected sample size of 15 and a probability of early termination of 0.736. The study was designed to study eltrombopag in ten patients in the first stage; the trial would be terminated if one or fewer achieved complete platelet response. Otherwise, the trial would go to the second stage, and 29 patients would be studied. If the total number of patients with complete platelet response were less than or equal to five, the drug would be deemed ineffective. The MF group was an exploratory group of ten patients to study the safety and activity of eltrombopag in patients with MF treated with ruxolitinib. We considered the activity promising if three or more patients out of ten achieved complete platelet response. For safety monitoring in the cohort with MF, accrual would stop if, at any time, four of ten patients encounter grade 3 or more nonhematological toxicity or progression to acute leukemia. As an additional safety procedure, we observed the first three MF patients on trial for at least 3 months before other patients were accrued. Response definitions Complete platelet response was defined as platelet count ≥50x109/L for CML, and ≥100x109/L for MF that was sustained for ≥3 months while continuing TKI or ruxolitinib therapy or with sustained (≥3 months) re-escalation of TKI dose without recurrence of thrombocytopenia. Criteria for CML and MF response were previously defined.21,22 The target response was a complete response in at least 30% of patients. Results Twenty-one patients were enrolled: 15 with CML and six with MF. Their median age was 60 years (range, 31-97) and their clinical characteristics are shown in Table 1. Median duration of disease was 2.2 years (range, 0.5-29 years) for patients with CML and 2 years (range, 0.3-3.6 years) for patients with MF. At the time of enrollment, patients with CML were receiving the following TKI: dasatinib (n=5), ponatinib (n=4), nilotinib (n=3), bosutinib (n=2), and imatinib (n=1), 27% were receiving their first TKI, 27% the second TKI, 27% the third, and 19% the fourth or later TKI. The median platelet count was 44x109/L (range, 3-49x109/L) in patients with CML and 62x109/L (range, 21-75x109/L) in those with MF. Cytogenetic response for patients with CML at baseline were partial in three, minor in six, and none in six. Prior therapies in MF patients were an investigational JAK2 inhibitor, and interferon a-2 in one patient each. The median dose of ruxolitinib was 10 mg (range, 10-30 mg) (Table 1). Eltrombopag dose distribution is summarized in Table 2. After a median duration of treatment of 18 months (range, 5-77 months), 12 of the 15 (80%) patients with CML achieved a complete platelet response with doses of eltrombopag of 50–300 mg per day. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). The median time to best response was 6 months (range, 2.1-13 months). Ten patients had sustained platelet recovery after stopping eltrombopag. The median duration for sustained platelet response was 45 months (range, 3-69 months). The three patients who did not achieve a complete platelet response had only minor changes in platelet count while they were taking eltrombopag (from 3x109/L to 8x109/L, 19x109/L to 45x109/L, and from 42x109/L to 46x109/L, respectively). Two patients (one each of CML and MF) had improvement in hemoglobin of over 2 g/dL from baseline (from 8.2 g/dL to 10.6 g/dL, and from 9.4 g/dL to 11.4 g/dL, respectively), Hemoglobin improvement was sustained over 21.5 and 2 months respectively while patients were taking eltrombopag. Hemoglobin levels declined after interruption of eltrombopag. One patient with CML had an absolute neutrophil count recovery to >1x109/L (baseline neutrophils 0.71x109/L). Absolute neutrophil count improvement was sustained for >6 months while on eltrombopag. Absolute neutrophil count then declined after interruption of eltrombopag. The TKI doses and duration for patients with CML post enrollment are summarized in Table 1. Nine patients with CML experienced an improvement in the cytogenetic response during the observation period (all of them had sustained platelet recovery after stopping eltrombopag); one from none to complete, two from minor to complete, four from minor to partial, and two from partial to complete (Table 3). In five patients with CML the TKI dose was increased and maintained while continuing eltrombopag. Dasatinib daily dose was increased from 50 mg to 100 mg in three patients, nilotinib dose was increased in one patient form 150 mg twice daily to 200 mg twice daily, and one patient had an increase in ponatinib dose from 15 mg every other day to 15 mg daily. There were no TKI dose-limiting toxicities in patients who increased their TKI doses. The dose increase was associated with improvement in CML response in four of these five patients. In the five CML patients who had a cytogenetic response but did not have TKI dose escalation, the improvement in cytogenetic response was noticed while patients were on eltrombopag. Three CML patients had a switch in their TKI (Online Supplementary Table S1). All three of these patients had already some improvement in thrombocytopenia before switching their TKI, with the change indicated for other non-hematologic adverse events in one patient and the inefficacy of the TKI in the other two patients. None of the six patients with MF responded (i.e., none had a sustained increase in platelet count to ≥100x109/L); minor upward transient variations in platelet counts were seen in three patients (from 21x109/L to 28x109/L, 41x109/L to 55x109/L and from 65x109L to 75x109/L, respectively). Table 1. Baseline characteristics. Table 2. Eltrombopag dose distribution, mg per day (all patients). As of the date of this report, 20 patients were off study because of a lack of response (n=9), stem cell transplant (n=2), death (n=2), patient’s wish (n=1), adverse events (n=2), TKI discontinuation (n=1), loss to follow-up (n=1) and stable platelets (n=2). The two deaths on study were not related to treatment. One death was secondary to infectious complication in a patient with MF. The second death was secondary to hemorrhagic shock in a CML patient. This patient, treated with dasatinib, developed hepatosplenomegaly and ascites while on study but the etiology was not known. There was no evidence of portal vein thrombosis on CT abdomen/pelvis. Both eltrombopag and dasatinib were held and she had no platelets response. The platelet count was 10x109/L at the time of death due to severe gastrointestinal and genitourinary bleeding. Figure 1. Platelet change from baseline to response. *Each blue bar reflects change in platelet count in a chronic myeloid leukemia patient, while each green bar reflects change in platelet count in a myelofibrosis patient. Therapy was well tolerated in most patients, but two patients on ponatinib developed thrombotic events. Two months after eltrombopag discontinuation due to termination of the study, one patient with CML developed significant thrombocytosis (>1,000x109/L) with a white blood cell count of 9.5x109/L, 3% basophils, and 2% peripheral blast accompanied by headache and eye pain. Ophthalmoscopic examination was suggestive of bilateral plaques or thrombosis in the retinal vasculature but fluoroscopic evaluation did not reveal retinal vasculature blockage. Ponatinib was discontinued and thrombocytosis was managed with hydroxyurea. The aforementioned symptoms resolved. There was no cytogenetic response prior or after starting eltrombopag. Seven months after stopping eltrombopag, the patient had a persistent increase in blasts to 13% without a lack of hematologic response and she was then started on a clinical trial with an investigational TKI. Another CML patient developed non-occlusive deep venous thrombosis in the right upper extremity without thrombocytosis while on ponatinib 4 months after the study was terminated. One MF patient who had a history of coronary artery disease status post coronary artery bypass surgery developed myocardial infarction (MI) while on eltrombopag. This patent had then worsening increase in bone marrow fibrosis from grade 2 to grade 3 and was taken off study 40 days after MI. No further additional thrombotic/thromboembolic complications in CML and MF patients observed during or after the study (Online Supplementary Table S2). One patient (CML) discontinued therapy due to toxicity (elevation of liver function tests). Grade 3/4 toxicities irrespective of attribution listed in Table 4. One patient with MF had an increase in bone marrow fibrosis from grade 2 to grade 3. That patient had an increase in blast from 3% to 8% in the peripheral blood and an increase from 1% to 6% in the bone marrow while he was on study but with an improvement in hemoglobin. There was no change in the patient's disease other than this change in blast percentage. The patient was taken off study for lack of platelet response and later started on another clinical trial (PRM-151 + ruxolitinib). No progression of disease has been documented in any other patients. No clonal evolution was observed in patients with prolonged eltrombopag use. Discussion Thrombocytopenia is a common adverse event in patients with CML and MF who are treated with TKI and ruxolitinib, respectively.10,23 In most instances, thrombocytopenia is transient, occurs early during treatment initiation, and can be successfully managed with transient treatment interruptions and occasionally dose adjustments. However, in some patients thrombocytopenia can be persistent and more severe requiring frequent treatment interruptions and dose reductions, which might adversely influence treatment outcome.11 To that end, rIL-11 was successfully used in CML patients for the management of TKI associated thrombocytopenia.16 The main limitation of use of rIL-11 in the management of chemotherapy-induced thrombocytopenia in solid malignancies was the narrow therapeutic window with significant fluid retention and occasional arrhythmias. However, at lower doses used in CML, it was well tolerated24,25 with grade 1 or 2 peripheral edema observed in six patients (43%). Eltrombopag is a second generation oral thrombopoietin receptor agonist that has induced improvement of thrombocytopenia in patients with immune-mediate thrombocytopenia (ITP) or aplastic anemia. The EXTEND trial demonstrated that long-term use of eltrombopag was effective in maintaining for more than 6 months platelet counts of 50×109/L or more and reducing bleeding in most patients with ITP. Addition of eltrombopag to immunosuppressive treatment also markedly increased overall and complete hematologic response rates in treatment-naive severe aplastic anemia.26 Here we describe the use of eltrombopag in the management of TKI-related thrombocytopenia in CML and MF. Our results suggest clinical benefit in most patients with CML with a generally favorable safety profile, although two patents (both on ponartinib) had thrombotic events. In contrast, no response was observed in patients with MF. Theoretical concerns about the use of eltrombopag in this setting include increase in marrow blasts and possible transformation to advanced phases, thrombotic events including portal vein thrombosis, and increase in marrow fibrosis. We did not observe any instance of transformation in our series, in concordance with pre-clinical and clinical data showing no evidence of worsening leukemia.20,27 There was also no increase in marrow fibrosis in CML patients. Our series is small so the lack of such events should be considered as preliminary but reassuring. The most common adverse event was LFT elevation, but these were generally transient, reversible and manageable with dose adjustments. However, in one case it led to discontinuation of eltrombopag because of recurrent transaminitis. Two patients who received ponatinib (50%) had thrombotic events while on eltrombopag, this might raise the precaution of using ponatinib in conjunction with eltrombopag in CML patients. Despite the median disease duration of 2.2 years and multiple TKI use in CML patients before enrollment, eltrombopag demonstrated clinical efficacy with complete platelet response of 80% (12 of 15). This compares favorably to what was reported with rIL-11.16 More important, nine patients (60%) had improvement in cytogenetic responses, likely the result of a more sustained therapy with TKI. Notably, as doses of eltrombopag were increased, LFT elevations were noted in some patients. Conversely, eltrombopag dose interruptions or reductions due to such events or to platelets reaching >200x109/L, occasionally resulted in a drop-in platelet counts. Thus, close monitoring and dynamic management is required, at least during the initial stages of therapy to obtain the maximum effect while maintaining safety. The lack of efficacy among patients with MF could be in part secondary to advanced disease, or possible antagonism between the two medications. Thrombopoietin agonist are dependent on JAK-stat pathway which is inhibited by ruxolitinib.28 Table 3. Response to eltrombopag in chronic myeloid leukemia patients. Table 4. Treatment emergent adverse events. Our study has several limitations. It was a small study, and it did not accrue to the target sample size of 29 patients due to slow enrollment making the observation preliminary and requiring confirmation. We also do not have evidence or investigation of any immune mechanisms associated with thrombocytopenia, although we believe it is unlikely that these patients with CML had an immune mediated thrombocytopenia, and uncommon occurrence in this setting. In conclusion, our findings show that eltrombopag doses up to 300 mg may alleviate TKI-associated thrombocytopenia in some patients with CML. No similar benefit has been observed in patients with MF treated with ruxolitinib. Although generally safe, thrombotic events were noted that deserve further investigation, particularly when used in combination with ponatinib. Additional studies are warranted to confirm these observations. Supplementary Material Supplementary Appendix
Fatal
ReactionOutcome
CC BY-NC
33054123
20,176,531
2021-11-01
What was the outcome of reaction 'Urogenital haemorrhage'?
The effect of eltrombopag in managing thrombocytopenia associated with tyrosine kinase therapy in patients with chronic myeloid leukemia and myelofibrosis. Approximately 20-50% patients with chronic phase chronic myeloid leukemia (CML-CP) treated with tyrosine kinase inhibitors (TKIs) or with myelofibrosis (MF) treated with ruxolitinib develop grade ≥3 thrombocytopenia needing treatment interruptions and dose reductions. We conducted a non-randomized, phase II, single-arm study to determine the efficacy of eltrombopag for patients with CML or MF with persistent thrombocytopenia while on TKI or ruxolitinib. Eltrombopag was initiated at 50 mg/day, with dose escalation up to 300 mg daily allowed every 2 weeks. Twenty-one patients were enrolled (CML=15, MF=6); median age 60 years (range, 31-97 years). The median platelet count was 44x109/L (range, 3-49x109/L) in CML and 62x109/L (range, 21-75x109/L) in MF. After a median of 18 months (range, 5-77 months), 12/15 patients with CML achieved complete platelet response. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). Among CML patients 5 could re-escalate the TKI dose and 9 improved their response. None of the 6 patients with MF had a sustained response. Therapy was generally well tolerated. One patient discontinued therapy due to toxicity (elevated transaminases). One patient with CML developed significant thrombocytosis (>1000x109/L). Another CML patient developed non occlusive deep venous thrombosis in the right upper extremity without thrombocytosis, and one MF patient had myocardial infarction. Eltrombopag may help improve platelet counts in CML patients receiving TKI with recurrent thrombocytopenia. Further studies are warranted. pmcIntroduction Tyrosine kinase inhibitors (TKI) are standard therapy for chronic myeloid leukemia (CML) and myelofibrosis (MF). Five TKI are currently approved for the treatment of CML in various stages, namely imatinib, nilotinib, dasatinib, bosutinib and ponatinib. Although these agents are generally well tolerated, some patients may develop adverse events, with myelosuppression being the most prominent.1-6 In most instances myelosuppression is grade 1 or 2 and requires no intervention. However, grade ≥3 thrombocytopenia (platelet ≤50x109/L) has been reported in 20% to 50% of patients. When this occurs, patients are usually managed with treatment interruption until platelets recovery (e.g., above 75x109/L) and dose reductions if thrombocytopenia recurs. Ruxolitinib is a JAK2 inhibitor used to manage splenomegaly and diseaseassociated symptoms in patients with MF.7 The dose limiting toxicity of ruxolitinib was thrombocytopenia8 and because of this the two pivotal phase III studies excluded patients with platelets ≤100x109/L. Still, thrombocytopenia was reported in 69% of patients, including 9% with grade ≥3.9 In patients with a platelet count of 50-100×109/L, grade ≥3 thrombocytopenia occurred in 56% of patients.10 Ruxolitinib-associated grade ≥3 thrombocytopenia is also typically managed with dose reductions or interruptions. Frequent dose interruptions and reductions might decrease TKI efficacy11,12 and may still not be sufficient to control thrombocytopenia. Patients who develop myelosuppression have a lower probability of achieving major or complete cytogenetic response (CCyR).11 For example, one study reported that patients treated with imatinib who developed grade ≥3 thrombocytopenia had a lower probability of CCyR compared to those who never developed thrombocytopenia (35% vs. 59%, P=0.02, respectively). Similarly, ruxolitinib efficacy is compromised with dose reductions and interruptions.12,13 In order to minimize dose reductions and interruptions, hematopoietic growth factors, filgrastim and erythropoietin stimulating agents (erythropoietin and darbepoetin) have been successfully used to manage neutropenia and anemia secondary to TKI in CML, respectively.14,15 Interleukin 11 (IL-11) was effective to manage thrombocytopenia associated with TKI in CML16 but use of this agent is associated with significant adverse events including fluid retention and cardiac arrhythmias. Eltrombopag is a non-peptide thrombopoietin receptor agonist that is effective and well tolerated among patients with immune thrombocytopenia, chronic hepatitis C-associated thrombocytopenia and severe aplastic anemia.17-19 It has also been safely used in acute myeloid leukemia without evidence of disease progression secondary to eltrombopag. 20 Here, we report the results from a pilot trial investigating the use of eltrombopag in the management of TKIor ruxolitinib-associated thrombocytopenia among patients with CML and MF. Methods Patients We conducted an open-label, non-randomized, phase II study of individualized dosing of eltrombopag. Eligible patients were aged 18 years or older with chronic phase CML receiving treatment with any Food and Drug Administration-approved TKI and experiencing grade ≥3 thrombocytopenia (platelets ≤50x109/L), or with MF receiving ruxolitinib and with platelets <100x109/L (since it is a dose-limiting toxicity and a label threshold for ruxolitinib), in either case after the first 3 months of therapy. Thrombocytopenia should have been either recurrent (i.e., be at least the second episode of thrombocytopenia) or have necessitated dose reductions of the TKI or ruxolitinib. All patients had signed an informed consent form approved by the Institutional Review Board, and the study was conducted in accordance with the Declaration of Helsinki. Study design Eltrombopag was commenced at 50 mg with dose escalation allowed every 2 weeks to 100 mg, 150 mg, 200 mg, and 300 mg (a higher dose than per label considering the thrombocytopenia refractoriness on these patients and the intent to continue TKI/ruxolitinib) according to platelet response. For patients of East Asian ancestry, eltrombopag was commenced at 25 mg daily with dose escalation allowed every 2 weeks. The following guideline was used to adjust dosing of eltrombopag: if the platelet count was >200x109/L at any time, the daily dose was reduced by 25 mg and re-assessed in 2 weeks; if >400x109/L, therapy was withheld and platelets assessed twice weekly until platelet count <150x109/L; therapy could then be resumed with the daily dose reduced by 25 mg. If the platelet count >400x109/L after 2 weeks was at the lowest dose, therapy was permanently discontinued. TKI doses were adjusted at the discretion of the treating physician per standard practice. Liver function tests (LFT) (alanine aminotransferase [ALT], aspartate aminotransferase [AST], and bilirubin) were done before the initiation of eltrombopag, every 2 weeks during the dose adjustment phase and following the monthly establishment of a stable dose. When LFT abnormalities were identified, LFT were performed weekly until the abnormalities resolved or stabilized; if ALT/AST levels were ≥ three-times the upper limit of normal (ULN): therapy was withheld, we then repeated abnormal liver function tests within 3-5 days; if confirmed abnormal, we monitored LFT weekly until resolved, stabilized, or returned to baseline. If ALT/AST levels ≥ three-times the ULN and were progressive, persistent (≥4 weeks), accompanied by increased direct bilirubin, or accompanied by clinical signs of liver injury or evidence of hepatic decompensation, eltrombopag was permanently discontinued. Patients who experienced other clinically significant grade 3 or greater toxicity possibly related to eltrombopag, had eltrombopag interruption until toxicity resolved to grade 1 or less. Treatment then was resumed at the immediate lower dose level. Failure to achieve a platelet count ≥50x109/L or ≥100x109/L in CML and MF patients, respectively after 8 weeks of eltrombopag was considered as lack of response. Statistical analysis Simon’s optimal two-stage design (Simon, 1989) was used to test the null hypothesis that the proportion of subjects with complete response is ≤0.10 versus the alternative that it is ≥0.30 (i.e., Po≤0.10 vs. Pa≥0.30) at alpha=0.05 with 80% power. The design resulted in an expected sample size of 15 and a probability of early termination of 0.736. The study was designed to study eltrombopag in ten patients in the first stage; the trial would be terminated if one or fewer achieved complete platelet response. Otherwise, the trial would go to the second stage, and 29 patients would be studied. If the total number of patients with complete platelet response were less than or equal to five, the drug would be deemed ineffective. The MF group was an exploratory group of ten patients to study the safety and activity of eltrombopag in patients with MF treated with ruxolitinib. We considered the activity promising if three or more patients out of ten achieved complete platelet response. For safety monitoring in the cohort with MF, accrual would stop if, at any time, four of ten patients encounter grade 3 or more nonhematological toxicity or progression to acute leukemia. As an additional safety procedure, we observed the first three MF patients on trial for at least 3 months before other patients were accrued. Response definitions Complete platelet response was defined as platelet count ≥50x109/L for CML, and ≥100x109/L for MF that was sustained for ≥3 months while continuing TKI or ruxolitinib therapy or with sustained (≥3 months) re-escalation of TKI dose without recurrence of thrombocytopenia. Criteria for CML and MF response were previously defined.21,22 The target response was a complete response in at least 30% of patients. Results Twenty-one patients were enrolled: 15 with CML and six with MF. Their median age was 60 years (range, 31-97) and their clinical characteristics are shown in Table 1. Median duration of disease was 2.2 years (range, 0.5-29 years) for patients with CML and 2 years (range, 0.3-3.6 years) for patients with MF. At the time of enrollment, patients with CML were receiving the following TKI: dasatinib (n=5), ponatinib (n=4), nilotinib (n=3), bosutinib (n=2), and imatinib (n=1), 27% were receiving their first TKI, 27% the second TKI, 27% the third, and 19% the fourth or later TKI. The median platelet count was 44x109/L (range, 3-49x109/L) in patients with CML and 62x109/L (range, 21-75x109/L) in those with MF. Cytogenetic response for patients with CML at baseline were partial in three, minor in six, and none in six. Prior therapies in MF patients were an investigational JAK2 inhibitor, and interferon a-2 in one patient each. The median dose of ruxolitinib was 10 mg (range, 10-30 mg) (Table 1). Eltrombopag dose distribution is summarized in Table 2. After a median duration of treatment of 18 months (range, 5-77 months), 12 of the 15 (80%) patients with CML achieved a complete platelet response with doses of eltrombopag of 50–300 mg per day. The median peak platelet count among responders was 154x109/L (range, 74-893x109/L). The median time to best response was 6 months (range, 2.1-13 months). Ten patients had sustained platelet recovery after stopping eltrombopag. The median duration for sustained platelet response was 45 months (range, 3-69 months). The three patients who did not achieve a complete platelet response had only minor changes in platelet count while they were taking eltrombopag (from 3x109/L to 8x109/L, 19x109/L to 45x109/L, and from 42x109/L to 46x109/L, respectively). Two patients (one each of CML and MF) had improvement in hemoglobin of over 2 g/dL from baseline (from 8.2 g/dL to 10.6 g/dL, and from 9.4 g/dL to 11.4 g/dL, respectively), Hemoglobin improvement was sustained over 21.5 and 2 months respectively while patients were taking eltrombopag. Hemoglobin levels declined after interruption of eltrombopag. One patient with CML had an absolute neutrophil count recovery to >1x109/L (baseline neutrophils 0.71x109/L). Absolute neutrophil count improvement was sustained for >6 months while on eltrombopag. Absolute neutrophil count then declined after interruption of eltrombopag. The TKI doses and duration for patients with CML post enrollment are summarized in Table 1. Nine patients with CML experienced an improvement in the cytogenetic response during the observation period (all of them had sustained platelet recovery after stopping eltrombopag); one from none to complete, two from minor to complete, four from minor to partial, and two from partial to complete (Table 3). In five patients with CML the TKI dose was increased and maintained while continuing eltrombopag. Dasatinib daily dose was increased from 50 mg to 100 mg in three patients, nilotinib dose was increased in one patient form 150 mg twice daily to 200 mg twice daily, and one patient had an increase in ponatinib dose from 15 mg every other day to 15 mg daily. There were no TKI dose-limiting toxicities in patients who increased their TKI doses. The dose increase was associated with improvement in CML response in four of these five patients. In the five CML patients who had a cytogenetic response but did not have TKI dose escalation, the improvement in cytogenetic response was noticed while patients were on eltrombopag. Three CML patients had a switch in their TKI (Online Supplementary Table S1). All three of these patients had already some improvement in thrombocytopenia before switching their TKI, with the change indicated for other non-hematologic adverse events in one patient and the inefficacy of the TKI in the other two patients. None of the six patients with MF responded (i.e., none had a sustained increase in platelet count to ≥100x109/L); minor upward transient variations in platelet counts were seen in three patients (from 21x109/L to 28x109/L, 41x109/L to 55x109/L and from 65x109L to 75x109/L, respectively). Table 1. Baseline characteristics. Table 2. Eltrombopag dose distribution, mg per day (all patients). As of the date of this report, 20 patients were off study because of a lack of response (n=9), stem cell transplant (n=2), death (n=2), patient’s wish (n=1), adverse events (n=2), TKI discontinuation (n=1), loss to follow-up (n=1) and stable platelets (n=2). The two deaths on study were not related to treatment. One death was secondary to infectious complication in a patient with MF. The second death was secondary to hemorrhagic shock in a CML patient. This patient, treated with dasatinib, developed hepatosplenomegaly and ascites while on study but the etiology was not known. There was no evidence of portal vein thrombosis on CT abdomen/pelvis. Both eltrombopag and dasatinib were held and she had no platelets response. The platelet count was 10x109/L at the time of death due to severe gastrointestinal and genitourinary bleeding. Figure 1. Platelet change from baseline to response. *Each blue bar reflects change in platelet count in a chronic myeloid leukemia patient, while each green bar reflects change in platelet count in a myelofibrosis patient. Therapy was well tolerated in most patients, but two patients on ponatinib developed thrombotic events. Two months after eltrombopag discontinuation due to termination of the study, one patient with CML developed significant thrombocytosis (>1,000x109/L) with a white blood cell count of 9.5x109/L, 3% basophils, and 2% peripheral blast accompanied by headache and eye pain. Ophthalmoscopic examination was suggestive of bilateral plaques or thrombosis in the retinal vasculature but fluoroscopic evaluation did not reveal retinal vasculature blockage. Ponatinib was discontinued and thrombocytosis was managed with hydroxyurea. The aforementioned symptoms resolved. There was no cytogenetic response prior or after starting eltrombopag. Seven months after stopping eltrombopag, the patient had a persistent increase in blasts to 13% without a lack of hematologic response and she was then started on a clinical trial with an investigational TKI. Another CML patient developed non-occlusive deep venous thrombosis in the right upper extremity without thrombocytosis while on ponatinib 4 months after the study was terminated. One MF patient who had a history of coronary artery disease status post coronary artery bypass surgery developed myocardial infarction (MI) while on eltrombopag. This patent had then worsening increase in bone marrow fibrosis from grade 2 to grade 3 and was taken off study 40 days after MI. No further additional thrombotic/thromboembolic complications in CML and MF patients observed during or after the study (Online Supplementary Table S2). One patient (CML) discontinued therapy due to toxicity (elevation of liver function tests). Grade 3/4 toxicities irrespective of attribution listed in Table 4. One patient with MF had an increase in bone marrow fibrosis from grade 2 to grade 3. That patient had an increase in blast from 3% to 8% in the peripheral blood and an increase from 1% to 6% in the bone marrow while he was on study but with an improvement in hemoglobin. There was no change in the patient's disease other than this change in blast percentage. The patient was taken off study for lack of platelet response and later started on another clinical trial (PRM-151 + ruxolitinib). No progression of disease has been documented in any other patients. No clonal evolution was observed in patients with prolonged eltrombopag use. Discussion Thrombocytopenia is a common adverse event in patients with CML and MF who are treated with TKI and ruxolitinib, respectively.10,23 In most instances, thrombocytopenia is transient, occurs early during treatment initiation, and can be successfully managed with transient treatment interruptions and occasionally dose adjustments. However, in some patients thrombocytopenia can be persistent and more severe requiring frequent treatment interruptions and dose reductions, which might adversely influence treatment outcome.11 To that end, rIL-11 was successfully used in CML patients for the management of TKI associated thrombocytopenia.16 The main limitation of use of rIL-11 in the management of chemotherapy-induced thrombocytopenia in solid malignancies was the narrow therapeutic window with significant fluid retention and occasional arrhythmias. However, at lower doses used in CML, it was well tolerated24,25 with grade 1 or 2 peripheral edema observed in six patients (43%). Eltrombopag is a second generation oral thrombopoietin receptor agonist that has induced improvement of thrombocytopenia in patients with immune-mediate thrombocytopenia (ITP) or aplastic anemia. The EXTEND trial demonstrated that long-term use of eltrombopag was effective in maintaining for more than 6 months platelet counts of 50×109/L or more and reducing bleeding in most patients with ITP. Addition of eltrombopag to immunosuppressive treatment also markedly increased overall and complete hematologic response rates in treatment-naive severe aplastic anemia.26 Here we describe the use of eltrombopag in the management of TKI-related thrombocytopenia in CML and MF. Our results suggest clinical benefit in most patients with CML with a generally favorable safety profile, although two patents (both on ponartinib) had thrombotic events. In contrast, no response was observed in patients with MF. Theoretical concerns about the use of eltrombopag in this setting include increase in marrow blasts and possible transformation to advanced phases, thrombotic events including portal vein thrombosis, and increase in marrow fibrosis. We did not observe any instance of transformation in our series, in concordance with pre-clinical and clinical data showing no evidence of worsening leukemia.20,27 There was also no increase in marrow fibrosis in CML patients. Our series is small so the lack of such events should be considered as preliminary but reassuring. The most common adverse event was LFT elevation, but these were generally transient, reversible and manageable with dose adjustments. However, in one case it led to discontinuation of eltrombopag because of recurrent transaminitis. Two patients who received ponatinib (50%) had thrombotic events while on eltrombopag, this might raise the precaution of using ponatinib in conjunction with eltrombopag in CML patients. Despite the median disease duration of 2.2 years and multiple TKI use in CML patients before enrollment, eltrombopag demonstrated clinical efficacy with complete platelet response of 80% (12 of 15). This compares favorably to what was reported with rIL-11.16 More important, nine patients (60%) had improvement in cytogenetic responses, likely the result of a more sustained therapy with TKI. Notably, as doses of eltrombopag were increased, LFT elevations were noted in some patients. Conversely, eltrombopag dose interruptions or reductions due to such events or to platelets reaching >200x109/L, occasionally resulted in a drop-in platelet counts. Thus, close monitoring and dynamic management is required, at least during the initial stages of therapy to obtain the maximum effect while maintaining safety. The lack of efficacy among patients with MF could be in part secondary to advanced disease, or possible antagonism between the two medications. Thrombopoietin agonist are dependent on JAK-stat pathway which is inhibited by ruxolitinib.28 Table 3. Response to eltrombopag in chronic myeloid leukemia patients. Table 4. Treatment emergent adverse events. Our study has several limitations. It was a small study, and it did not accrue to the target sample size of 29 patients due to slow enrollment making the observation preliminary and requiring confirmation. We also do not have evidence or investigation of any immune mechanisms associated with thrombocytopenia, although we believe it is unlikely that these patients with CML had an immune mediated thrombocytopenia, and uncommon occurrence in this setting. In conclusion, our findings show that eltrombopag doses up to 300 mg may alleviate TKI-associated thrombocytopenia in some patients with CML. No similar benefit has been observed in patients with MF treated with ruxolitinib. Although generally safe, thrombotic events were noted that deserve further investigation, particularly when used in combination with ponatinib. Additional studies are warranted to confirm these observations. Supplementary Material Supplementary Appendix
Fatal
ReactionOutcome
CC BY-NC
33054123
20,176,531
2021-11-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Drug ineffective for unapproved indication'.
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
AMLODIPINE BESYLATE, CARBAMAZEPINE, DROXIDOPA, FLUDROCORTISONE, METHYLPREDNISOLONE, MIDODRINE, PYRIDOSTIGMINE
DrugsGivenReaction
CC BY-NC-ND
33055478
20,836,871
2021-09-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Off label use'.
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
AMLODIPINE BESYLATE, CARBAMAZEPINE, DROXIDOPA, FLUDROCORTISONE, METHYLPREDNISOLONE, MIDODRINE, PYRIDOSTIGMINE
DrugsGivenReaction
CC BY-NC-ND
33055478
20,836,871
2021-09-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Orthostatic hypotension'.
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
AMLODIPINE BESYLATE, CARBAMAZEPINE, DROXIDOPA, FLUDROCORTISONE, METHYLPREDNISOLONE, MIDODRINE, PYRIDOSTIGMINE
DrugsGivenReaction
CC BY-NC-ND
33055478
20,836,871
2021-09-15
What was the dosage of drug 'CARBAMAZEPINE'?
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
400 MG
DrugDosageText
CC BY-NC-ND
33055478
20,836,871
2021-09-15
What was the dosage of drug 'DROXIDOPA'?
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
900 MG, QD
DrugDosageText
CC BY-NC-ND
33055478
20,836,871
2021-09-15
What was the dosage of drug 'FLUDROCORTISONE'?
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
0.2 MG, QD
DrugDosageText
CC BY-NC-ND
33055478
20,836,871
2021-09-15
What was the dosage of drug 'METHYLPREDNISOLONE'?
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
1000 MG, QD
DrugDosageText
CC BY-NC-ND
33055478
20,836,871
2021-09-15
What was the dosage of drug 'MIDODRINE'?
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
6 MG, QD
DrugDosageText
CC BY-NC-ND
33055478
20,836,871
2021-09-15
What was the dosage of drug 'PYRIDOSTIGMINE'?
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
120 MG, QD
DrugDosageText
CC BY-NC-ND
33055478
20,836,871
2021-09-15
What was the outcome of reaction 'Orthostatic hypotension'?
Anti-LGI1 Encephalitis Developing Immunoglobulin Responsive Orthostatic Hypotension after Remission. Anti-leucine-rich glioma-inactivated 1 (LGI1) antibody is associated with limbic encephalitis. We herein report a patient with anti-LGI1 encephalitis who developed severe orthostatic hypotension (OH) responsive to immunoglobulin therapy five years after developing symptoms of encephalitis. A 71-year-old man presented with amnesia caused by limbic encephalitis. The symptoms of encephalitis improved partially without any immunotherapy. Five years later, he developed severe OH, and anti-LGI1 antibody was positive. The catecholamine dynamics indicated that the central autonomic nervous system was the lesion of his OH. Intravenous immunoglobulin therapy improved the OH. This case suggests that anti-LGI1 antibody can be associated with severe OH. pmcIntroduction Anti-voltage-gated potassium channel (VGKC) complex antibody-associated syndrome contains a wide spectrum of neurological diseases, such as limbic encephalitis, Morvan's syndrome, and neuromyotonia (1). The antibodies include mainly anti-contactin-associated protein-like 2 (CASPR2) antibody and anti-leucine-rich glioma-inactivated 1 (LGI1) antibody, and the clinical manifestations differ depending on the antibody. Anti-LGI1 antibody is commonly associated with limbic encephalitis, which usually causes subacute memory deficit, behavioral and spatial disorientation, seizure, and hyponatremia (1). However, orthostatic hypotension (OH) has not been reported as a symptom. We herein report a patient with anti-LGI1 encephalitis who developed severe OH responsive to immunoglobulin therapy about five years later. Case Report A 71-year-old man was admitted to our department because of memory disturbance. He had previously been healthy but developed amnesia three months before admission. Upon admission, he exhibited a severe impairment of his recent memory and an irritable mood. His intelligence was preserved, and he did not show any other neurological symptoms or seizure. His Mini-Mental State Examination (MMSE) score was 25/30, and delayed recall was impaired (1/3). His delayed recall score on the Wechsler Memory Scale-Revised (WMS-R) was 56. Brain magnetic resonance imaging (MRI) showed left-dominant medial temporal lobe signal hyperintensity on fluid-attenuated inversion recovery (FLAIR) without gadolinium enhancement (Fig. 1a). These areas exhibited increased blood flow on N-isopropyl-p-[123I] iodoamphetamine single-photon emission computed tomography (123IMP-SPECT) (Fig. 1b). An electroencephalogram (EEG) showed transient theta bursts at the bilateral frontal lobes. Serum sodium was 135 mEq/L (136.0-145.0 mEq/L). A cerebrospinal fluid analysis showed that protein (37.1 mg/dL) and cell counts (2/μL) were within the normal ranges. Through the examination, prostate cancer was found. Radiation and hormone therapies were initiated for prostate cancer, and the serum level of prostate specific antigen (PSA) decreased from 11.5 ng/mL to below 1.0 ng/mL (normal range 0.0-4.0 ng/mL). We prescribed 400 mg of carbamazepine for the abnormal EEG. His mood was stabilized, and the memory disturbance remained without further deterioration. Although his serum turned out to be positive for the anti-LGI1 antibody, we did not perform any immunotherapies. Figure 1. Brain imaging. Radiological findings of encephalitis (a, b) and five years after remission (c, d). During encephalitis, MRI showed FLAIR hyperintensity at the bilateral medial temporal lobes (a), and 123IMP-SPECT showed an increase in blood flow at the left medial temporal lobe (b). MRI five years after remission showed diminished signal abnormality and slight atrophy of the affected area (c). No increased blood flow was observed on 123IMP-SPECT (d). FLAIR: fluid-attenuated inversion recovery, 123IMP-SPECT: N-isopropyl-p-[123I] iodoamphetamine single photon emission computed tomography, MRI: magnetic resonance imaging At 76 years old, 5 years later, the patient complained of dizziness when standing up and developed hypertension (systolic blood pressure: about 200 mmHg). He was prescribed 2.5 mg of amlodipine and visited our hospital when he became unable to stand up by himself a week later. Upon admission, he had mild amnesia, which had not changed in five years. He did not show any other neurological symptoms. His MMSE score was 26/30, with a delayed recall score of 3/3. His delayed recall score on the WMS-R was still 56. The Schellong test showed severe OH; his systolic blood pressure was over 140 mmHg in the supine position. However, his radial artery pulsation became impalpable in the upright position, accompanied by a feeling of fainting. We ceased the administration of amlodipine, but his OH did not improve. He had diarrhea and constipation. Routine laboratory examinations showed no apparent abnormalities. A cerebrospinal fluid analysis showed a slightly elevated protein level (51.6 mg/dL), normal cell count (2/μL), and a negative oligoclonal band. The serum anti-LGI1 antibody was positive, whereas the anti-CASPR2 antibody was negative. Antibodies for the following were negative: serum anti-ganglionic acetylcholine (ACh) receptor, anti-neural antibodies (amphiphysin, CV-2, PNMA2, Ri, Yo, Hu, recoverin, SOX1, titin, zic4, GAD65, Tr), and cerebrospinal fluid anti-N-methyl-D-aspartate receptor (NMDAR). The serum PSA level was 0.8 ng/mL. The patient was still on carbamazepine, and epileptic discharge was not observed on an EEG. An electrocardiogram showed a heart rate of 70 bpm with a normal sinus rhythm. The coefficient of variation of the R-R interval (CVR-R) was slightly decreased (1.6%). On brain MRI, the bilateral medial temporal lobes had shrunk and the hyperintensity lesions diminished (Fig. 1c). The blood flow decreased in the same area on 123IMP-SPECT (Fig. 1d). 123I-ioflupane SPECT showed a normal uptake in both striata. 123I-metaiodobenzylguanidine (MIBG) myocardial scintigraphy was normal (early and delayed H/M ratios of 2.73 and 2.75, respectively, and washout rate of 30.5%). 18F-fluorodeoxyglucose positron emission tomography showed no abnormal uptake in the whole body, including the prostate. His OH was not improved by midodrine (6 mg/day), droxidopa (900 mg/day), pyridostigmine (120 mg/day), or fludrocortisone (0.2 mg/day). Three courses of methylprednisolone therapy (1,000 mg/day, three days) did not relieve his symptoms. Three weeks after the last methylprednisolone therapy, we administered intravenous immunoglobulin (IVIg) (400 mg/kg/day, 5 days). His blood pressure gradually stabilized, and he became able to walk without any assistance two weeks after the IVIg administration. However, eight weeks after the infusion of IVIg, he developed dizziness and became unable to stand by himself. His OH relapsed, so he was re-admitted to our hospital. Immunoglobulin therapy improved his symptoms two weeks after the second IVIg administration. We conducted a head-up tilt test before and after IVIg treatment (Fig. 2). In the resting position, the blood pressure, noradrenaline (NA), and arginine vasopressin (AVP) values were within normal limits. However, when raising his head, his blood pressure dropped remarkably, and he felt nauseous and nearly fainted. His NA and AVP levels increased as the blood pressure dropped. When we moved him back to the flat position, rebound hypertension was observed. After the treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The amount of increase in the NA and AVP levels was larger than that before the treatment. Figure 2. The tilt test before and after intravenous immunoglobulin (IVIg) therapy. The patient was placed in the supine position for 15 min before the tilt test. The head side of the table was raised every 5 min to 15°, 30°, 45°, 60°, and 80°, and then restored to the horizontal position. To analyze NA and AVP, blood samples were obtained every 5 min just before changing positions and 10 min after restoring the flat position. In the resting position, the BP, NA, and AVP values were within normal limits. However, before treatment and while raising the head, his BP dropped remarkably, and he felt nauseous and nearly fainted; the NA and AVP levels increased as the BP decreased. When he returned to the flat position, rebound hypertension was observed. After treatment, his systolic BP did not decrease below 80 mmHg, and he did not feel nauseous. The increases in the NA and AVP levels were larger than those before treatment. AVP: arginine vasopressin, BP: blood pressure, HR: heart rate, IVIg: intravenous immunoglobulin, NA: noradrenaline Discussion We described a case of anti-LGI1 encephalitis which developed severe OH responsive to IVIg five years after the symptoms of encephalitis. Anti-LGI1 encephalitis can manifest with subacute memory deficit, behavioral and spatial disorientation, seizures, and hyponatremia. Patients develop combinations of these symptoms (1). Our patient developed subacute memory deficit and an irritable mood. Although he did not manifest with seizure or hyponatremia, his clinical course was compatible with anti-LGI1 encephalitis. We conducted treatment for the prostate cancer, considering the encephalitis to be a paraneoplastic manifestation of the prostate cancer. After chemotherapy for the prostate cancer was initiated, his mood stabilized, and his cognitive dysfunction stopped deteriorating without any immunotherapy. The increased blood flow on 123IMP-SPECT had also disappeared. However, spontaneous recovery of anti-LGI1 encephalitis has been reported (2). Furthermore, there is only one report of the concurrence of prostate cancer and anti-LGI1 encephalitis (3). Accordingly, the encephalitis might have improved as part of its natural course. Based on the clinical assessments, we concluded that the OH of our patient was due to immune-mediated central autonomic dysfunction. MIBG scintigraphy showed a normal uptake, suggesting that the OH had not been caused by peripheral autonomic dysfunction. The NA and AVP values at rest were within normal limits and increased when we raised the patient's head (Fig. 2). These results suggest that his OH was caused by a disturbance of the central efferent pathway of the autonomic nervous system, including the hypothalamus and medulla oblongata (Table) (5). Before presenting with severe OH, the patient developed hypertension, possibly as a result of sympathetic overactivity syndrome. The improvement in his OH by the administration of IVIg suggested an autoimmune mechanism underlay his OH. Table. Relation between Cathecolamine Dynamics during the Tilt Test and the Lesions of Orthostatic Hypotension. Resting AVP ΔAVP Resting NA ΔNA Peripheral afferent normal low normal-high low Peripheral efferent normal-high preserved low low Central afferent normal low normal preserved Central efferent normal-high preserved various value preserved AVP: arginine vasopressin, NA: noradrenaline Modified from Zerbe RL, et al. Am JMed 1983; 74: 265-271 OH can be caused by several conditions, such as drug use, Parkinson's disease, multiple system atrophy, polyneuropathy, and autonomic autoimmune ganglionopathy (AAG) (5). The adverse effects of drugs, neurodegenerative disorders, or neuropathy were excluded by the clinical assessment. Although AAG is an autoimmune disorder that causes autonomic dysfunction responsive to IVIg, it was considered unlikely because it causes postganglionic autonomic failure, and nearly half of AAG cases are positive for anti-ganglionic ACh receptor antibody (6). In some types of encephalitis, such as anti-NMDAR encephalitis and anti-GAD encephalitis, autonomic dysfunction can be induced by the impairment of the subthalamus or brainstem (7-9). However, these antibodies were negative. Since the above-mentioned known causes of OH were excluded, we suspected that the OH was associated with anti-LGI1 antibody. Anti-LGI1 antibody binds to the magnocellular neurons of the paraventricular nucleus (PVN) of the subthalamus and causes dysregulated ADH secretion, which is considered the cause of hyponatremia in anti-LGI1 encephalitis (10). The PVN also includes the parvocellular neurons, which project to autonomic preganglionic neurons in the spinal cord (11). When anti-LGI1 antibody binds to the parvocellular neurons of the PVN, dysregulation of the blood pressure is expected to occur. Anti-LGI1 antibody inhibits the ligand-receptor interaction between LGI1 and ADAM22/23, resulting in a reduction in synaptic AMPA receptors (12). Some studies have shown that AMPA receptors in the PVN play an important role in central blood pressure regulation (13,14). Furthermore, the PVN is involved in the central efferent pathway of the autonomic system, which corresponded to the supposed lesion of the OH in our patient. Our patient likely lacked accompanying seizure because he was taking carbamazepine, which had been prescribed for his irritable mood. The patient did not experience any worsening of his recent memory in the interim, possibly because he had already developed memory disturbance and no further disturbance was evident at that time. We cannot exclude the possibility that unknown neuronal surface antibodies, such as the other isotype of anti-VGKC antibody, were involved in the OH in the present patient. The association between anti-LGI1 antibody and OH is unclear for several reasons. First, we were unable to investigate the correlation between the antibody titer and the clinical course. Second, there was a temporal dissociation between OH and the symptoms of encephalitis. Third, there have been no other reports describing the concurrence of anti-LGI1 antibody and OH. The present findings suggest that anti-LGI1 antibody may be associated with severe OH. However, unknown antibodies may have caused the OH in our patient. The further accumulation of similar patients is thus warranted to elucidate the pathophysiology of immunotherapy-responsive OH. The authors state that they have no Conflict of Interest (COI). Acknowledgement We thank Dr. Akihiro Mukaino and Prof. Shunya Nakane from the Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto for screening for the anti-ganglionic acetylcholine receptor antibody.
Recovering
ReactionOutcome
CC BY-NC-ND
33055478
20,836,871
2021-09-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Blood corticotrophin decreased'.
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
AMLODIPINE BESYLATE, DROSPIRENONE\ETHINYL ESTRADIOL, HYDROCORTISONE, METYRAPONE
DrugsGivenReaction
CC BY-NC-ND
33055479
19,217,578
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Cortisol increased'.
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
AMLODIPINE BESYLATE, DROSPIRENONE\ETHINYL ESTRADIOL, HYDROCORTISONE, METYRAPONE
DrugsGivenReaction
CC BY-NC-ND
33055479
19,217,578
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Drug interaction'.
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
AMLODIPINE BESYLATE, DROSPIRENONE\ETHINYL ESTRADIOL, HYDROCORTISONE, METYRAPONE
DrugsGivenReaction
CC BY-NC-ND
33055479
19,217,578
2021-03-15
What was the administration route of drug 'DROSPIRENONE\ETHINYL ESTRADIOL'?
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
Oral
DrugAdministrationRoute
CC BY-NC-ND
33055479
19,217,578
2021-03-15
What was the administration route of drug 'HYDROCORTISONE'?
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
Oral
DrugAdministrationRoute
CC BY-NC-ND
33055479
19,217,578
2021-03-15
What was the dosage of drug 'DROSPIRENONE\ETHINYL ESTRADIOL'?
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
3/0.02MG DAILY
DrugDosageText
CC BY-NC-ND
33055479
19,217,578
2021-03-15
What was the dosage of drug 'HYDROCORTISONE'?
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
15?20MG DAILY
DrugDosageText
CC BY-NC-ND
33055479
19,217,578
2021-03-15
What was the outcome of reaction 'Blood corticotrophin decreased'?
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
Recovered
ReactionOutcome
CC BY-NC-ND
33055479
19,217,578
2021-03-15
What was the outcome of reaction 'Cortisol increased'?
Oral Contraceptive Disturbed the Recovery of the Adrenal Function after Adrenalectomy in Cushing Syndrome. Estrogen is known to increase exogenous corticosteroid levels. In this case, a 27-year-old Japanese woman was referred to our hospital for examination of an adrenal tumor and was diagnosed with Cushing syndrome. Resection of the tumor resulted in secondary adrenal insufficiency. She also developed microcytic anemia due to hypermenorrhea, which was masked by Cushing syndrome. An oral contraceptive was administered for the treatment of hypermenorrhea, but this led to a marked increase in serum cortisol and the reduction of plasma adenocorticotropic hormone, disturbing the recovery of the adrenal function. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Introduction If patients originally have hypermenorrhea, it may be masked by Cushing syndrome, which causes amenorrhea. In such cases, the masked hypermenorrhea may become apparent when hypercortisolism is cured. While oral contraceptives are generally used for prevention of hypermenorrhea, the administration of estrogen is known to increase endogenous or exogenous corticosteroids (1-6). We herein present a case in which the recovery of the adrenal function was disturbed by oral contraceptive use after adrenalectomy for Cushing syndrome. Attention is required when oral contraceptives are used to treat hypermenorrhea masked by Cushing syndrome. Case Report A 27-year-old Japanese woman with an adrenal tumor was referred to our hospital for an endocrinological assessment. One year previously, her blood pressure had been 200/100 mmHg and treatment with amlodipine (5 mg per day) was initiated. Since then, she noticed general fatigue, moon face, red striae, acne and menstrual irregularity. Abdominal MRI revealed right adrenal mass of 3 cm in diameter, which was isointense to liver on T2-weighted sequences. On abdominal CT, the tumor was homogenous and exhibited low attenuation values (2.3 HU). On admission, the plasma adenocorticotropic hormone (ACTH) was <1.0 pg/mL and the serum cortisol level was 23.6 μg/dL. The serum potassium level was 3.2 mEq/L. The urinary cortisol level was 150.8 μg/day. The serum cortisol level after a 1-mg dexamethasone suppression test (DST) was 23.7 μg/dL and the midnight serum cortisol level was 20.8 μg/dL. The renin-aldosterone ratio and urinary catecholamine levels were within the normal range. She was diagnosed with adrenal Cushing syndrome. Hypercortisolism was managed by block and replacement therapy using metyrapone (1,500 mg per day) and hydrocortisone (15-20 mg per day) for three months. Then the tumor was laparoscopically resected and was histologically diagnosed as adrenal cortical adenoma. After resection of the tumor, she developed secondary adrenal insufficiency and was treated with oral hydrocortisone. The time course of her adrenal function after adrenalectomy is shown in Figure. At all points, blood samples were collected before 10 AM without the morning dose of hydrocortisone. At all points, except for 15 months after adrenalectomy, hydrocortisone was administered in the evening on the day before blood collection. From 5 to 12 months after adrenalectomy, her plasma ACTH and serum cortisol levels gradually recovered to 33 pg/mL and 1.5 μg/dL, respectively. Although her hemoglobin (Hb) level was normal (12.3 g/dL) before adrenalectomy, she had microcytic anemia (Hb 10.2 g/dL and mean cell volume 78.8 fL) at that time, most likely due to hypermenorrhea after adrenalectomy. One month later (13 months after adrenalectomy), she consulted a gynecologist and was treated with YazⓇ [Drospirenone (3 mg per day) and ethinyl estradiol (0.02 mg per day)] for hypermenorrhea. At the next visit (14 months after adrenalectomy), her serum cortisol was markedly elevated to 19.9 μg/dL, whereas the plasma ACTH level dropped to <1 pg/mL. Her anemia normalized. One month later (15 months after adrenalectomy), when she skipped the evening dose of hydrocortisone on the day before blood collection, her serum cortisol level was reasonably low at 0.6 μg/dL. Three months later (18 months after adrenalectomy) when the evening dose of hydrocortisone was administered the day before blood collection, her serum cortisol was again found to be high (5.9 μg/dL) whereas her plasma ACTH level was suppressed to 1 pg/ml. As oral contraceptives are reported to increase circulating free cortisol levels after exogenous hydrocortisone administration (6), we suspected that YazⓇ might have impaired the disappearance of the evening dose of hydrocortisone, and the administration of YazⓇ was stopped. Two months later (20 months after adrenalectomy), her serum cortisol was reasonably low (0.7 μg/dL) and her plasma ACTH level had slightly recovered to 6 pg/mL. To facilitate the recovery of the adrenal function, the evening dose of hydrocortisone was stopped and her plasma cortisol and ACTH levels were gradually normalized until 41 months after adrenalectomy, although a rapid ACTH test indicated partial impairment of the adrenal function (serum cortisol, 12.2 μg/dL) at 31 months after adrenalectomy. Figure. The time course of the plasma ACTH and serum cortisol levels after adrenalectomy. From 13 to 18 months after adrenalectomy, an oral contraceptive was administered. Blood was collected before 10 a.m., without the administration of the morning dose of hydrocortisone. The evening dose of hydrocortisone was administered on the day before blood collection at every point, except for 15 months after adrenalectomy, when the evening dose of hydrocortisone was skipped the day before blood collection (*). Discussion While the recovery of the adrenal function from secondary adrenal insufficiency requires appropriate oral hydrocortisone dosing, various studies have demonstrated the effects of estrogen on circulating corticosteroid levels. Even low-dose estrogen increases the endogenous total and free cortisol levels (1-4, 7) and reduces the plasma ACTH level, possibly through a negative feedback mechanism (7, 8). Similarly, the action of exogenous hydrocortisone is enhanced by relatively high-dose estrogen (approximately equivalent to 15-120 mg of diethylstilbestrol) (5), but low-dose oral contraceptives also increase circulating free cortisol levels after exogenous hydrocortisone administration (6). These effects of estrogen on free cortisol were considered to be mediated by the concomitant increase in hepatic corticosteroid-binding globulin (CBG) synthesis (1-4). CBG is the main reservoir of cortisol in the human body, binding 70-80% of total cortisol. The CBG-bound cortisol is protected from disappearance in the blood, which results in increased free cortisol, as well as the increase in the total cortisol concentration and the ratio of total to free cortisol (9, 10). In this case, the patient's original hypermenorrhea was concealed by amenorrhea in Cushing syndrome. With the cure of Cushing syndrome, the masked hypermenorrhea became apparent and was treated with an oral contraceptive. With the constant dose of hydrocortisone, the oral contraceptive increased the total (and possibly free) serum cortisol levels and suppressed the plasma ACTH level, probably disturbing the recovery of the adrenal function. In fact, the adrenal function was still partially impaired at 31 months after adrenalectomy, which is longer than one year, which is the average time to recovery from adrenal Cushing's syndrome after adrenalectomy (11). These hypotheses would have been verified by the measurement of CBG during and after oral contraceptive treatment. Instead of oral contraceptives, oral progestin may be considered for the treatment of hypermenorrhea, as progesterone has little effect on the metabolism of cortisol (12). Clinicians should be careful of this potential pitfall in the management of secondary adrenal insufficiency after Cushing syndrome. The authors state that they have no Conflict of Interest (COI).
Recovered
ReactionOutcome
CC BY-NC-ND
33055479
19,217,578
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Delirium'.
Impact of Geriatric Pharmacy Specialist Interventions to Reduce Potentially Inappropriate Medication Among Hospitalized Elderly Patients at Medical Wards: A Prospective Quasi-Experimental Study. BACKGROUND Elderly patients are at greater risk of receiving potentially inappropriate medications (PIMs) and developing adverse drug events. Identification and correction of PIMs is essential to maximize medication safety. OBJECTIVE To determine the prevalence of PIMs on admission in Thai elderly patients admitted to a medical ward and to compare changes of PIMs on discharge, following comprehensive care by a ward pharmacist with or without a geriatric pharmacy specialist. METHODS A prospective, quasi-experimental study was performed at a tertiary university hospital in Bangkok, Thailand. Patients aged ≥ 60 years who were admitted to the medical ward were recruited and allocated to one of two groups: intervention (IG) and control (CG). The CG received pharmaceutical care from the ward pharmacist. The IG received pharmaceutical care from the geriatric pharmacy specialist along with the ward pharmacist. The 2012 Beers criteria were used to identify PIMs on admission, during hospitalization, and on discharge. RESULTS Prevalence of PIMs on admission was 43.3% (N = 187). On discharge, prevalence of PIMs in the IG decreased significantly compared to that on admission (21.3% and 43.3%, p < 0.05) and was significantly lower than in the CG (21.3% and 40.9%, p = 0.036). Moreover, the percentage of patients without PIMs on discharge in the IG was significantly higher than in the CG (78.7% and 59.1%, p < 0.0001). CONCLUSIONS Use of PIMs was common among hospitalized elderly patients on admission. Pharmaceutical care provided by a geriatric pharmacy specialist in conjunction with a ward pharmacist significantly reduced the prevalence of PIMs on discharge compared with on admission. Introduction Many countries, including Thailand, have a rapidly increasing elderly population. An older population is seven times more likely to be hospitalized due to adverse drug events (ADEs) than a younger population [1, 2]. Common risk factors for ADEs in this population include polypharmacy, inappropriate prescribing, and pharmacokinetic and pharmacodynamic changes [3, 4]. The majority of adverse drug reactions (ADRs) in older people are ‘type A reactions,’ which are otherwise preventable. In a prospective study of 1,756 admitted patients aged over 65 years, 45.1% of ADRs were classified as definitely avoidable and 31.4% as potentially avoidable [1]. There are several strategies to further reduce this ADE rate, one of which is the identification of potentially inappropriate medications (PIMs). PIM is defined as a medication for which the potential risk for ADE is higher than the supposed clinical benefit. Dispensing PIMs in the elderly also constitutes suboptimal prescribing, especially if a safer alternative is available. Therefore, several prescribing criteria have been developed to minimize prescribing errors in elderly patients [5–9]. The most referred prescribing criteria, Beers criteria, were developed by the American Geriatric Association based on the expert opinions of geriatric pharmacotherapy specialists [10]. In elderly patients, up to 16% of medications being taken at the time of admission are defined as PIMs, and almost 50% of admitted patients have at least one PIM [1]. Thus, the identification and management of PIMs might be the best way to prevent ADE in the elderly. Several studies, focused on the prevalence of PIMs using Beers criteria, have shown that around 42.6% of prescribed medications were PIMs [11–16]. However, most of these studies were conducted in outpatient settings. The few studies with inpatient settings have demonstrated a different rate of PIMs (around 50–70%) [12]. Furthermore, a recent meta-analysis found that multi-disciplinary teams involving pharmacists can reduce PIMs by reducing prescribing errors in the older population [13, 14]. Some studies focused on the prevalence of PIMs in elderly patients in Thailand [15], but most of them boasted a retrospective, cross-sectional descriptive design in an outpatient setting; very few studies were carried out at the inpatient unit in primary- or secondary-care hospitals. In addition, these studies used different explicit criteria including Beers criteria and newly developed criteria [16–18]. Therefore, the objective of this study was to determine the prevalence of PIMs in hospitalized elderly Thai patients. In addition, this study aimed to compare the reduction in PIMs on discharge between patients receiving pharmaceutical care delivered by a geriatric pharmacy specialist and patients receiving the usual care by a ward pharmacist. Methods Study Design, Setting, and Patient Population This was a prospective, quasi-experimental, single-center study conducted at Siriraj Hospital, Bangkok, Thailand from May 2015 to February 2016. All patients aged 60 years or older who were admitted to these medical wards and provided informed consent were included in the study. Patients were excluded if they were admitted for less than 48 h, transferred to other wards, died during admission, or needed either palliative or end-of-life care, as determined by the physician. The study protocol was approved by the review committee of Faculty of Medicine Siriraj Hospital, Mahidol University. Concerning the characteristic of medical wards in our setting, they consisted of eight medical wards with 20 beds in each ward. These wards had been divided into an intervention group (four wards) and a control group (four wards), therefore we expected that there would be 80 patients in each group at the same time. Regarding the criteria for admission, patients were admitted to a vacant bed in each ward assigned by the routine hospital admission protocol; the investigators were not involved in this procedure. Eligible patients were allocated into either an intervention or a control group depending on their admitted ward. One inpatient ward pharmacist worked regularly with the medical team in each medical ward and one geriatric pharmacy specialist, with a board certified geriatric pharmacotherapy credential, worked among four wards of the intervention group. In the intervention group, the patients were provided pharmaceutical care by the geriatric pharmacy specialist adjunct to the ward pharmacist. In the control group, the patients received standard pharmaceutical care by the ward pharmacist. The standard care included medication reconciliation within 48 h of patient admission, drug use evaluation, providing drug information to the medical team, and discharge counseling. In addition, the geriatric pharmacy specialist in the intervention group assessed the appropriateness of medication during hospitalization every day, using the 2012 Beers criteria to identify any PIMs and provided a suitable intervention to the physician to optimize drug therapy based on age-related physiological changes and their clinical significance. The team communicated verbally in person or by written notes on the patient chart. Outcome Measurements The primary outcome was the prevalence of PIMs on admission. Secondary outcomes included changes in the proportion of PIMs on discharge compared with that on admission between the two groups and changes in each type of PIMs in each group on discharge. PIMs were identified according to the 2012 Beers criteria by the same geriatric pharmacy specialist throughout the study for both groups. In the intervention group, PIMs were detected via a medical chart review, within 48 h after admission, every day during hospitalization, and on discharge. In the control group, however, the PIMs were identified retrospectively by a medical chart review after the patients were discharged. The identified PIMs in both groups were classified into three subtypes for subgroup analysis: general PIMs, condition-associated PIMs, and PIMs with anticholinergic properties. Furthermore, all patients in both groups were followed up via phone call by the geriatric pharmacy specialist at 2 weeks after discharge to assess medication adherence, possible adverse events, and other potential medical and drug-related problems. If drug-related problems such as side effects were identified, the geriatric pharmacy specialist provided solutions to the patients and their caregivers. However, if a serious medical illness was found, rehospitalization or an outpatient visit was recommended. Sample Size Calculation and Statistical Analysis The sample size was calculated based on the results obtained from Egger et al. [19], which revealed that the estimated prevalence of PIMs in hospitalized elderly patients was 0.16. Thus, at least 207 patients had to be recruited for a significance level of α = 0.05 and a power of at least 80% (β < 0.20). All data were tested for normal distribution by measuring kurtosis and skewness. The baseline characteristics were compared between groups by using Fisher’s exact test for nominal data and the Mann–Whitney U test for continuous data. The primary analytical approach was a per-protocol analysis. Descriptive analysis was applied for the number of PIMs. The χ2 test was used to compare the prevalence of PIMs between groups on admission, during hospitalization, and on discharge. A paired t test was used to compare the prevalence of PIMs on discharge with that on admission in the same group. McNemar’s test was used to test the nominal data. A p value of less than 0.05 was considered to indicate a statistically significant difference. All analyses were performed using the Statistical Package for the Social Sciences for Windows version 21. Results Baseline Characteristics In total, 234 patients were enrolled, of whom 47 patients (20%) were excluded according to the exclusion criteria. Of these, two were admitted for less than 48 h, three were transferred to other wards, 39 died during admission, and three needed either palliative or end-of-life care, as determined by the physician. Then, the 187 eligible patients were divided into two groups with 94 and 93 patients in the intervention and control groups, respectively. There was no significant difference between the baseline characteristics of the patients in both groups (Table 1). The mean age of the patients in the intervention and control group was 74.0 ± 8.4 and 74.9 ± 9.0 years, respectively. Both groups had more than 50% male population and their functional status could be identified as partially dependent. The average number of prior medications per patient on admission was 7.8 ± 4.4 and 8.7 ± 5.1 items in the intervention and control groups, respectively. The most common causes of hospitalization for both groups were heart failure, pneumonia, non-ST segment elevation myocardial infarction, chronic obstructive pulmonary disease with acute exacerbation, and upper gastrointestinal bleeding.Table 1 Baseline characteristics of patients in the intervention group and control group (N = 187) Characteristic Intervention group (n = 94) Control group (n = 93) p value Age (years), mean ± SD  60–74 years, n (%)  75–84 years, n (%)  ≥ 85 years, n (%) 74.0 ± 8.4 57 (60.6) 24 (25.5) 13 (13.8) 74.7 ± 9.0 46 (49.5) 30 (32.3) 17 (18.3) 0.567 Sex: male, n (%) BMI (kg/m2), mean ± SD 48 (51.1) 21.9 ± 3.9 48 (51.6) 22.58 ± 4.0 0.940 0.220 Functional status 0.207  Independent, n (%)  Partially dependent, n (%)  Totally dependent, n (%) 29 (30.9) 41 (43.6) 24 (25.5) 20 (21.5) 52 (55.9) 21 (22.6) Cause of admission, n (%) 0.157  Heart failure  Pneumonia  NSTEMI  COPD with AE  UGIB 11 (11.7) 15 (16) 6 (6.4) 5 (5.3) 6 (6.4) 12 (12.9) 7 (7.5) 6 (6.5) 5 (5.4) 3 (3.2) Average number of medications prior to admission, mean ± SD (mode) 7.8 ± 4.4 (8) 8.7 ± 5.1 (9) 0.250 Length of hospital stay (days), mean, median (range) 16.97, 14 (3–78) 13.41, 10 (3–51) 0.084 BMI body mass index, COPD with AE chronic obstructive pulmonary disease with acute exacerbation, NSTEMI non-ST segment elevation myocardial infarction, UGIB upper gastrointestinal bleeding Prevalence of Potentially Inappropriate Medications (PIMs) We found that 81 patients took at least one PIM on admission; thus, the overall prevalence of PIMs on admission was 43.3% (intervention group: 43.6%, control group: 43.0%), with no significant difference between the groups (p = 0.636) (Table 2). The most common PIM classes are summarized in Table 3.Table 2 Prevalence of potentially inappropriate medications (PIMs) during the study Admission (%) Hospitalization (%) Discharge (%) Total (N = 187) 43.3 46.0 31.0* Intervention group (n = 94) 43.6 43.6 21.3** Control group (n = 93) 43.0 48.4 40.9 *p < 0.05 compared between the prevalence of PIMs on discharge and on admission **p = 0.036 compared between intervention group and control group on discharge Table 3 Potentially inappropriate medication (PIM) classes on admission (N = 187) Medication/medication class Percentage Benzodiazepines 21.70 Doxazosin 16.04 Orphenadrine + paracetamol 7.55 Amitriptyline 6.60 NSAIDs 5.66 Hydroxyzine 5.66 Trihexyphenidyl 4.72 Cyproheptadine 3.77 Methyldopa 2.83 Megestrol 2.83 NSAIDs non-steroidal anti-inflammatory drugs The prevalence of PIMs during hospitalization slightly increased but had no significant difference when compared with the prevalence of PIMs on admission in both groups. On discharge, the overall prevalence of PIMs was found to be significantly decreased from that on admission (31.0% and 43.3%, respectively, p < 0.05). Moreover, the prevalence of PIMs in the intervention group was found to be significantly lower than that in the control group (21.3% and 40.9%, respectively, p = 0.036). Consequently, the percentage of patients without PIMs in the intervention group was significantly higher than that in the control group (78.7% and 59.1%, respectively, p < 0.0001; Fig. 1).Fig. 1 Percentage of patients without potentially inappropriate medications (PIMs) on admission, during hospitalization and on discharge Types of PIMs Through the Hospital Course On admission, the most prevalent type of PIMs found in both groups was general PIMs (56.7% vs. 57.4% in the intervention and control group, respectively; Fig. 2). The prevalence of condition-associated PIMs and PIMs with anticholinergic properties in the intervention group was found to be 10.0% and 33.3%, respectively. In contrast, in the control group, the prevalence of PIMs was found to be 14.8% and 27.9%, respectively. Overall, there was no significant difference in the prevalence of each type of PIM between groups.Fig. 2 Prevalence of potentially inappropriate medications (PIMs) in the intervention group and control group on admission, during hospitalization, and on discharge. PIM_int PIMs in intervention group, PIM _ctrl PIMs in control group. *Significant difference between on admission and discharge (p < 0.05) During hospitalization, the prevalence of general PIMs and condition-associated PIMs slightly increased in both groups (Fig. 2). In contrast, the prevalence of PIMs with anticholinergic properties declined in both groups. However, the prevalence of each type of PIM showed no statistical difference when compared with that on admission. On discharge, the prevalence of general PIMs in the intervention group was found to be significantly decreased when compared with that on admission (39.3% vs. 56.7%, respectively, p < 0.0001). In contrast, this prevalence in the control group showed no difference from that on admission (58.5% vs. 57.4%, respectively). The prevalence of condition-associated PIMs increased during hospitalization in both groups but did not show any statistically significant difference when compared with that on admission. The prevalence of PIMs with anticholinergic properties did not show much difference in the intervention group from that on admission, while it decreased in the control group from that on admission, but without a significant difference. All patients found to be prescribed PIMs on discharge in the intervention group (n = 20) and the control group (n = 38) were scheduled to have a health assessment by phone call 2 weeks after being discharged from the hospital. The results showed that some patients in both groups experienced adverse events, such as dizziness, sedation, and headache, with no need for medication management (Table 4). Only three patients in the intervention group and six patients in the control group developed serious adverse events, which needed medical attention. One patient in the intervention group developed delirium from anticholinergic usage (cyproheptadine) and one patient in the control group experienced confusion and disorientation, due to a suspected acute fever, requiring rehospitalization. Another patient in the control group with a history of myocardial infarction developed acute heart failure after taking non-steroidal anti-inflammatory drugs (NSAIDs) obtained from a drug store and required rehospitalization.Table 4 Clinical outcomes at 2 weeks after discharge from hospital in patients who had potentially inappropriate medications (PIMs) on discharge Number of patients Intervention group (n = 94) Control group (n = 93) Patients with no PIMs on discharge 74 55 Patient had at least one PIM on discharge 20 38 Average number of PIMs on discharge (mean ± SD) 1.25 ± 0.77 1.32 ± 0.62  No adverse events 9 (45.0%) 14 (36.8%)  Developed adverse events but no need for other treatment 7 (35.0%) 16 (42.1%)  Developed adverse events and need medication management 3 (15.0%) 6 (15.8%)  Rehospitalization 1 (5.0%) 2 (5.3%) Discussion All patients enrolled in the study were allocated to the wards by the inpatient unit officer, depending on their physician and availability of units in each ward. Therefore, this study had a quasi-experimental design, as true randomization was not possible. In addition, each medical ward at the study site was operated separately. We ensured that the ward pharmacist provided identical pharmaceutical care in each ward every day that complied with the standard care of pharmacy professionals. However, the results from our unpublished preliminary observational study showed that PIMs were still detected in the prescription of elderly patients who were discharged from these wards. Consequently, a geriatric pharmacy specialist with expert knowledge and experience in geriatric pharmacotherapy could help to fill the gap in standard care delivered by the ward pharmacist in the medical ward [20]. Therefore, this may be the first study to verify the impact of geriatric pharmacy specialist involvement on the prevalence of PIMs in hospitalized elderly patients. In our study, we used 2012 Beers criteria to identify PIMs. Thai healthcare professionals, including physicians and pharmacists, are more familiar with the Beers criteria than others. Thus, lack of knowledge about medication lists to be avoided in the elderly could not be given as a reason. Furthermore, the ward pharmacist informed the relevant physician about the appropriateness of medication by referring to the Beers criteria for elderly patients. Accordingly, we could assess the impact of the geriatric pharmacy specialist on the administration of PIMs during hospitalization when compared with the impact of ward pharmacist in the control group. This may also explain the reduction of PIMs in the control group during hospitalization and on discharge, especially PIMs with anticholinergic properties. Data from 187 eligible patients were analyzed, and this number was lower than the expected sample size (N = 207). This figure was not used to determine the impact of interventions between intervention and control groups. The results of our study demonstrated that PIMs could be detected during the study period in both groups. Therefore, the reduced number of participants should not have affected to the overall results. Regarding PIMs detection, we found that 43.3% of our patients were taking at least one PIM on admission. This finding is comparable with that of previous studies, where the prevalence of PIMs in hospitalized patients ranged between 44.4% and 79.0% [21, 22], and this range of values is higher than that in the outpatient setting (16.0–53.0%) [6, 23–25]. Our study could not be considered a pioneer study in terms of studying the prevalence of PIMs in Thailand. However, previous studies that have assessed PIMs in elderly Thai patients have been mostly performed in an outpatient setting [15, 26]. The reported prevalence of PIMs was in the range of 19.2% and 28.1%, which is relatively smaller than that in our study [15]. Moreover, the PIMs on admission in our study were calculated from the lists of medications prior to admission, and should not have differed from those in outpatient settings. However, the comparison with other studies must be interpreted carefully to account for differences in the patient settings, criteria used to detect PIMs, and the study methodology employed. The most commonly observed PIMs in our study were benzodiazepines, such as diazepam, lorazepam, clonazepam, and NSAIDs, comparable to other studies [6, 27]. However, our results also showed doxazosin, an alpha-1 receptor antagonist, as a common PIM, which diverged from previous study results. Doxazosin is the treatment of choice for symptomatic benign prostatic hypertrophy, the most common prostate problem for older men [28]. As half of our eligible participants were male (51%) and over 60 years old, they were likely to receive doxazosin for this indication. However, doxazosin has a high risk of orthostatic hypotension and is not recommended as a routine treatment for patients with hypertension [29]. In addition, a safer alpha-1 receptor antagonist with highly selective and subtype-specific, such as tamsulosin, is available, which is generally better tolerated than other alpha-1 receptor antagonists (terazosin, prazosin, doxazosin) and has no clinically relevant effect on blood pressure [29, 30]. With regards to the prevalence of PIMs on admission between groups, no statistically significant difference in the prevalence of PIMs, and the number of general PIMs, condition-associated PIMs, and PIMs with anticholinergic properties was observed between the study groups. This might be related to the similarity in the baseline characteristics between the intervention and control groups, particularly the number of medications on admission. During hospitalization, the prevalence of total PIMs in both groups counted cumulatively across the hospital stay did not show a significant difference when compared with that on admission. However, there was a relatively higher prevalence of general PIMs and condition-associated PIMs in both groups than that on admission. Notably, the oral long-acting benzodiazepines, NSAIDs, and medication with anticholinergic properties, which were commonly identified PIMs on admission, had been discontinued on the recommendation of the ward pharmacist after completion of medication review and reconciliation. In addition, the geriatric pharmacy specialist recommended discontinuation of long-acting sulfonylurea (glyburide) and central alpha-1 receptor agonist (methyldopa) in the intervention group. The ward pharmacist and the physician may have been unaware of these medications being PIMs. Nevertheless, some new general PIMs were documented during hospitalization, especially at night time, when neither the ward pharmacist nor geriatric pharmacy specialist worked at the ward. Examples of these PIMs were meperidine injection for severe acute pain, oral lorazepam for insomnia and anxiety, oral metoclopramide for nausea, and intravenous diazepam for status epilepticus. Noticeably, most of these medications have safer alternatives except for status epilepticus [31, 32]. Similarly, using oral short- to intermediate-acting benzodiazepine for short-term symptomatic treatment of acute anxiety (not more than 2 weeks) was considered appropriate [33]. We believe that these recognized PIMs were continued during hospitalization and at discharge because the physician considered that their benefit outweighed their short-term risk. Condition-associated PIMs was determined according to a list of medications that should be avoided in older adults with certain diseases. The increased number of condition-associated PIMs in the intervention group was related to antipsychotics. Antipsychotics should be avoided in patients with dementia or cognitive impairment because they can increase the risk of cerebrovascular events. In our study, dementia was found in 4.3% and 3.3% of the patients in the intervention and control groups, respectively. Thus, antipsychotics, such as risperidone, quetiapine, and olanzapine, were used to treat behavioral and psychological symptoms of dementia such as agitation and delusion which usually flared up with stress, change in daily routines, excessive stimulation and change in environment. Accordingly, these antipsychotic agents were prescribed during hospitalization and extended until discharge. In contrast, the PIMs with anticholinergic properties during hospitalization were satisfactorily decreased in both groups when compared with on admission. This may be a result of the healthcare professionals’ familiarity with the Beers criteria, which recommend avoiding anticholinergic drugs in elderly patients. Therefore, medications with anticholinergic properties found during the medication review (such as hydroxyzine, orphenadrine, and trihexyphenidyl) were recognized by the physician and the ward pharmacist and generally discontinued if there was no specific therapeutic indication. On the other hand, PIMs with anticholinergic properties on discharge were associated with the increasing use of loratadine, which was a non-sedating antihistamine for treatment of allergic symptoms. Loratadine was classified as a PIM with anticholinergic properties according to the 2012 Beers criteria. Nevertheless, it is removed from the list in updated 2015 Beers criteria. Similarly, on discharge, the prevalence of PIMs in the intervention group was significantly lower than that on admission, whereas only a tiny reduction of total PIMs in the control group was observed when compared with that on admission. Correspondingly, a statistically significant difference in the percentage of patients without PIMs on discharge was observed between the two groups. To our knowledge, PIMs are associated with an increased risk of poor outcomes in the older population including adverse events, drug–drug or drug–disease interaction, and hospitalization [34, 35]. Accordingly, the patients in the intervention group had better outcomes compared with those in the control group, owing to medication safety. Regardless, there were no established data to demonstrate the relationship between the presence of PIMs on discharge and the risk of adverse effects or rehospitalization rate [36–39]. In our study, the telephone follow-up at 2 weeks post-discharge by the geriatric pharmacy specialist revealed that most of the patients were healthy, despite having PIMs on discharge, while some patients developed adverse events despite having no PIMs on discharge. This finding indicated that, in addition to identifying PIMs to assess medication safety, the management of individualized pharmacokinetic and pharmacodynamic changes, renal function, hepatic function, co-morbidities, and concurrent medications providing recommendations for dose adjustment and discharge counseling plan should also be included in the care protocol to maximize the safety of medication use. Regarding the prevalence of PIMs subtype on discharge in the intervention group, there was a 31% reduction of general PIMs compared with that on admission. Although there was an observed increase in general PIMs during hospitalization as discussed above, these new PIMs were prescribed as a once-a-day regimen. Thus, these identified PIMs were withdrawn on discharge. In addition, other medications accounting for general PIMs had been discontinued since admission. Therefore, the prevalence of general PIMs on discharge was found to be dramatically decreased. In contrast, no significant changes in the prevalence of condition-associated PIMs and PIMs with anticholinergic properties on discharge were observed compared with on admission, despite intervention by the geriatric pharmacy specialist. This non-acceptance of pharmacist interventions might be a result of the inpatient healthcare physician’s incomplete understanding of the patient’s previous illness and medication history. Furthermore, some PIMs were prescribed by specialists or senior physicians, which inpatient physicians are often reluctant to modify or stop even pharmacist’s recommendation. In summary, our study results disclosed that including a geriatric pharmacy specialist in the medical team had an impact on the prevalence of PIMs in hospitalized elderly patients, specifically general PIMs. In addition, our study also emphasized the importance of having a ward pharmacist in the medical team, as they help in complete medication review and reconciliation, including modification or discontinuation of PIMs in the hospitalized elderly patients, especially PIMs with anticholinergic properties. Nevertheless, other healthcare professionals including physicians and nurses may help to diminish PIMs as well, provided that they are trained on rational drug use in the elderly. The presence of a pharmacist as a drug expert in the medical team will, however, help reinforce the optimization of drug therapies for elderly patients. Additionally, providing the ward pharmacist with a short training course related to geriatric pharmacotherapy might improve their competency and reduce the prevalence of PIMs, resembling the impact made by a geriatric pharmacy specialist. There are some obvious limitations to our study. First, several enrolled patients were excluded from the study, and this number was more than that anticipated. This may be related to our study setting being the biggest university hospital in Bangkok. Thus, majority of the patients admitted to our hospital, with co-morbidities and moderate-to-severe disease severity, had to be excluded. Second, we could not assess whether the adverse events that occurred after discharge were correlated with the residual PIMs at discharge. Third, the PIMs identification in the control group was carried out retrospectively after the patient was discharged. Consequently, medication administration records or data related to drug therapy were unavailable or incomplete, thus, such PIMs could not be counted and the prevalence of PIMs could be underestimated. Conclusion The Beers-related PIMs use was highly prevalent among Thai patients aged over 60 years who were hospitalized in the medical ward. Applied geriatric pharmacotherapy knowledge and pharmaceutical skills delivered by a geriatric pharmacy specialist together with the ward pharmacist has the potential to reduce the prevalence of PIMs in this population, as demonstrated by significantly less PIMs on discharge than that on admission. The general PIMs and PIMs with anticholinergic properties are the most common subtypes that can be optimized. However, a shortage of ward pharmacists, especially geriatric pharmacy specialists, may pose a big challenge for the implementation of this model in the general inpatient setting. Acknowledgements The authors would like to express their great appreciation to all participants, physicians, nurses, ward pharmacists, and all medical staffs at the study wards and those who helped them to complete this study. Declarations Funding Not applicable. Conflict of interest The authors declare that there are no conflicts of interest. Ethics approval The study protocol was approved by the review committee of Faculty of Medicine Siriraj Hospital, Mahidol University. Consent to participate Signed consent forms were obtained from all eligible patients. Consent for publication Not applicable. Availability of data and material A subsection on data described in the manuscript, including all relevant raw data, will be freely available to any researcher wishing to use them for non-commercial purposes, without breaching participant confidentiality. Code availability Not applicable. Author contributions Conceptualization: WC, VS, TS; methodology: WC, VS, TS; formal analysis and investigation: WC, VS, TS; writing—original draft preparation: WC, TS; writing—review and editing: TS; resources: VS, TS; supervision: VS, TS.
CYPROHEPTADINE
DrugsGivenReaction
CC BY-NC
33063296
19,406,939
2021-03
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Stress fracture'.
Clinical and Radiological Characterization of Patients with Immobilizing and Progressive Stress Fractures in Methotrexate Osteopathy. Methotrexate (MTX) is one of the most commonly prescribed drugs for autoimmune rheumatic diseases. As there is no consensus on its negative effects on bone, the purpose of this investigation was to determine the clinical spectrum of patients with stress fractures due to long-term MTX treatment (i.e., MTX osteopathy). We have retrospectively analyzed data from 34 patients with MTX treatment, severe lower extremity pain and immobilization. MRI scans, bone turnover markers, bone mineral density (DXA) and bone microarchitecture (HR-pQCT) were evaluated. Stress fractures were also imaged with cone beam CT. While the time between clinical onset and diagnosis was prolonged (17.4 ± 8.6 months), the stress fractures had a pathognomonic appearance (i.e., band-/meander-shaped, along the growth plate) and were diagnosed in the distal tibia (53%), the calcaneus (53%), around the knee (62%) and at multiple sites (68%). Skeletal deterioration was expressed by osteoporosis (62%) along with dissociation of low bone formation and increased bone resorption. MTX treatment was discontinued in 27/34 patients, and a combined denosumab-teriparatide treatment initiated. Ten patients re-evaluated at follow-up (2.6 ± 1.5 years) had improved clinically in terms of successful remobilization. Taken together, our findings provide the first in-depth skeletal characterization of patients with pathognomonic stress fractures after long-term MTX treatment. Introduction Methotrexate (MTX) is a folate antagonist that is used in low doses (5–25 mg/week) in the first-line treatment of rheumatoid arthritis (RA), as well as in other inflammatory diseases such as systemic lupus erythematosus (SLE). It acts by inhibiting dihydrofolate reductase, which is an essential factor in DNA and RNA synthesis, but also has far-reaching anti-inflammatory and immunoregulatory effects [1, 2]. While the effects of MTX on bone metabolism have not been completely elucidated, there is both clinical and in vitro evidence for its adverse effects on bone. MTX osteopathy was first described in 1984 in children who had undergone prolonged maintenance therapy with oral MTX due to acute lymphocytic leukemia (ALL) [3], where distal femoral and tibial fractures with thick dense provisional zones of calcification were detected. In the 1990s, several clinical reports described “MTX osteopathy”, including low bone mass and stress fractures of the distal and proximal tibiae [4–6]. Since then, stress fractures of the proximal tibia have also been described in other patients after long-term MTX use [7, 8]. Inhibitory effects of MTX on osteoblastic bone formation have been detected in human biopsies as well as in murine bone cells [4, 9, 10], and bone mechanical properties were impaired in MTX-treated rats [11]. Although the documented effects of MTX include stimulation of both pro-inflammatory and anti-inflammatory pathways, increased cytokine production may be a possible mechanism for tissue damage in certain conditions such as in MTX osteopathy. In this regard, MTX treatment in cell culture has been linked to a dose-dependent increase in pro-inflammatory cytokines such as IL-1 and IL-6 [12]. At higher doses, MTX use led to increased TNF-α levels and promoted osteoclastogenesis [13]. MTX osteopathy has also been questioned based on the observation that MTX treatment in small patient cohorts and after short-term follow-up did not result in changes in bone turnover or bone density [14], and comparable bone loss compared to other disease-modifying antirheumatic drugs (DMARDs) [15]. Therefore, the clinical relevance of MTX osteopathy remains unclear. In general, the impairment of bone mineral density and quality as well as the increased risk of fracture have been reported in a variety of rheumatic diseases; however, they are often attributed to inflammatory processes and corticosteroid therapy [16–18]. As there are indications of low bone formation and elevated bone resorption in MTX osteopathy, a combination of anti-resorptive (i.e., denosumab) and osteoanabolic (i.e., teriparatide) therapy might be the most logical treatment option to improve bone strength and recover from stress fractures. We have previously demonstrated recovery from stress fractures in an SLE patient with MTX osteopathy after MTX discontinuation and combined denosumab–teriparatide treatment [19]. Here, we present a clinical characterization of 34 patients suffering from stress fractures after long-term methotrexate use for different underlying rheumatologic diseases and outline their positive response to denosumab and teriparatide treatment in a subset of ten patients. Methods Subjects Thirty-four patients with long-term (> 3 years) methotrexate use and stress fractures were included in this study. All patient data were evaluated in a retrospective and anonymized design. Data were analyzed according to the rules of the local ethics committee of the University Medical Center Hamburg-Eppendorf, Germany. Skeletal Assessment and Imaging Studies While magnetic resonance imaging (MRI) was performed for individual stress fracture detection, we also determined the areal bone mineral density (aBMD) using dual-energy X-ray absorptiometry (Lunar iDXA; GE Healthcare; Madison, WI, USA) and bone turnover markers from serum and urine samples in all patients. The measured serum markers included 25-hydroxyvitamin D (25-OH-D3), parathyroid hormone (PTH), osteocalcin (Oc), bone-specific alkaline phosphatase (BAP), and deoxypyridinoline cross-links in the urine (DPD). Furthermore, bone microarchitecture was analyzed in 30/34 patients using high-resolution peripheral quantitative computed tomography (HR-pQCT; XtremeCT, Scanco Medical, Switzerland). In the remaining four patients, HR-pQCT could not be performed due to bilateral distal tibia fractures. Scans were performed in the nonfractured distal tibia following a standardized procedure using the standard in vivo patient evaluation protocol [20, 21]. Specifically, we analyzed the trabecular bone mineral density (Tb.BMD), trabecular number (Tb.N), trabecular thickness (Tb.Th) and cortical bone mineral density (Ct.BMD). Stress fractures were also imaged in ten patients using cone beam computed tomography (CBCT) working at 90 kV, 40 mAs with a field of view of 16 × 16 ×  13 cm and slice thickness of 0.2 mm (SCS MedSeries H22, Planmed Oy, Helsinki, Finland). CBCT is a novel imaging technique using divergent X-rays that form a cone, which is increasingly used in extremity imaging, though its initial application was in dentistry and maxillofacial surgery [22, 23]. CBCT enables better fracture detection at extremity sites compared to standard radiography [24]. Furthermore, the spatial resolution of CBCT is higher and the radiation dose exposure is lower compared to conventional multislice CT [25]. Bone Biopsy Studies We obtained four bone biopsies from individuals with long-term MTX treatment and stress fractures. The first biopsy was obtained from an 81-year-old woman suffering from a stress fracture of the proximal right tibia. The biopsy was obtained from the lateral (unaffected) part of the tibial plateau in the course of total knee arthroplasty (TKA) and compared to a biopsy obtained from a 79-year-old female patient with primary osteoarthritis undergoing TKA. The other three biopsies were obtained from fracture sites as a part of a diagnostic work-up in these patients. While the second biopsy was obtained from the distal tibia of a 64-year-old patient previously reported elsewhere [19], the third and fourth biopsies were taken from the distal tibia and the calcaneus of a 71-year-old woman with RA. The specimens were fixed in 3.7% formaldehyde, dehydrated, embedded in methyl-methacrylate, and cut on a Microtec rotation microtome (CVT 4060E, Micro Tec, Walldorf, Germany). Afterwards the 5-µm sections were stained with toluidine blue and von Kossa. Histomorphometric analysis was performed according to the ASBMR nomenclature committee [26]. BV/TV, Tb.N, and Tb.Th as well as osteoid volume per bone volume (OV/BV) were evaluated in the von Kossa-stained sections. The osteoblast surface per bone surface (Ob.S/BS) and the osteoclast surface per bone surface (Oc.S/BS) were evaluated from the toluidine blue-stained sections. Quantitative backscattered electron imaging (qBEI) was performed on the first biopsy using a scanning electron microscope (LEO 435 VP; LEO Electron Microscopy Ltd., Cambridge, England) with a backscattered electron detector (Type 202; K.E. Developments Ltd., Cambridge, England), as described previously [27]. The scanning electron microscope was operated at 20 kV and 680 pA at a constant working distance. The acquired images were analyzed using a customized MATLAB (The MathWorks, Inc., Natick, Massachusetts, USA) script. QBEI was used to measure the bone mineral density distribution based on the generated gray values that represent the mean calcium content (mean Ca-Wt%). For both samples, calcium distribution curves were calculated. Treatment Intervention and Follow-Up Ten patients were evaluated at a follow-up visit at 2.6 ± 1.5 years (min. 1 year, max. 5 years). MTX treatment had been discontinued in 8/10 patients in consultation with a rheumatologist, while the MTX dose was reduced in the remaining 2 patients. Seven out of 10 patients were treated with a subcutaneous administration of denosumab 60 mg (Prolia®, Amgen, USA) every 6 months in combination with a daily subcutaneous administration of teriparatide 20 µg (Forsteo®, Eli Lilly, USA). While two patients were not treated with this bone-specific therapy due to individual contraindications, one patient was treated with teriparatide only. The two patients with no bone-specific treatment had discontinued their MTX medication. All patients received 20,000 IE vitamin D3 and 1 g of dietary calcium daily. At follow-up, clinical reexamination and an HR-pQCT scan were performed. The clinical course was evaluated, and patients were asked about subjective changes in mobility and pain (++/+ major/minor improvement, −/− no improvement/worsened). Furthermore, we performed the Timed Up and Go test and compared the achieved time to previously published reference values [28]. The chair rising test (CRT) was performed as described previously [29], and the results were compared to reference data [30]. Statistical Analysis All data were evaluated using GraphPad Prism® (GraphPad Software, La Jolla, CA, USA). Data are presented as scatter plots with additional labeling of the mean value ± the standard deviation (SD). After checking for normal distribution, the paired t-test was used to compare measurement results from the initial presentation and follow-up. Furthermore, the percent change per year was calculated. P-values of 0.05 or less were considered statistically significant. Results Clinical Characterization A total of 34 patients were evaluated, of whom all had load-dependent pain of the lower limb without adequate trauma in the past. MRI pointed to band- or meander-shaped stress fractures paralleling the former provisional zones of calcification and growth plates in all cases. The most frequent location was the distal tibia (53%) and the calcaneus (53%), followed by the proximal tibia (44%) and the distal femur (18%). In 68% of the patients, we observed stress fractures at multiple and/or bilateral locations (Fig. 1a–e). No association between the time interval of clinical onset and diagnosis and the MTX treatment durations or doses could be detected.Fig. 1 MRI morphology of stress fractures in MTX osteopathy. Band-like stress fractures in proton density (PD)-weighted fat-suppressed (FS) turbo spin echo (TSE) MRI sequences are seen. a Distal tibia, coronal and sagittal plane of two different patients. b Calcaneus, coronal and sagittal plane of two different patients. c Proximal tibia/distal femur, coronal plane. d Multiple stress fractures were found in 68% of the patients. e Bar graph indicating the regional distribution of stress fractures in n = 34 patients (frequency in % for each skeletal site) MTX osteopathy was diagnosed mostly in females (88%, Fig. 2a). The MTX dosage was 18.6 ± 4.9 mg (15–25 mg) weekly. Importantly, a prolonged time between clinical onset and diagnosis of 17.4 ± 8.6 months (5–36 months) was noted. Most patients had a history of corticosteroid treatment; however, upon presentation at our clinic, only 12/34 patients (35.3%) received low-dose oral prednisone (< 5 mg) (Fig. 2b). MTX treatment had been prescribed due to RA (26/34 patients), psoriatic arthritis (4/34 patients), SLE (2/34 patients), polymyalgia rheumatica (1/34 patients) or ankylosing spondylitis (1/34 patients) (Fig. 1c). Only two patients had vertebral fractures. We observed a peak in the occurrence of MTX osteopathy at the age of 70–79 years (Fig. 2d).Fig. 2 Clinical characteristics and bone mineral density, turnover and microstructure. a Eighty-eight percent of the affected patients were female. b Prednisone/no prednisone treatment. c Distribution of the different rheumatic diseases. d Age distribution. e, f DXA T-score at the lumbar spine (LS) and total hip. g, h Serum bone-specific alkaline phosphatase (BAP) and osteocalcin levels (both bone formation) and i urinary deoxypyridinoline (DPD) (bone resorption). Gray boxes indicate reference ranges. j Trabecular number (Tb. N), k trabecular thickness (Tb. Th) and l cortical BMD compared to age- and sex-matched reference data [31] A total of 21/34 (61.8%) of the patients were diagnosed with osteoporosis (i.e., BMD T-score ≤ − 2.5), and the remaining patients were diagnosed with osteopenia, according to the DXA measurements (Fig. 2e, f). Biochemical bone turnover analyses revealed higher levels in the bone-specific alkaline phosphatase than in the osteocalcin levels compared to reference values (Fig. 2g, h). In 18/32 patients (56%), we detected elevated bone resorption markers in the urine (i.e., DPD cross links) (Fig. 2i). HR-pQCT analysis revealed an almost normal trabecular number in most patients and a more severe reduction of trabecular thickness and cortical BMD in the distal tibia compared to reference values (Fig. 2j–l). While two patients presented with 25-hydroxy-vitamin D3 levels < 20 µg/l, ten patients had 25-OH-D3 levels < 30 µg/l. All patients received oral vitamin D supplementation. Cone Beam CT (CBCT) Imaging and Biopsy Findings CBCT imaging demonstrated osseous alterations around the typical stress fracture localizations (distal tibia, around the knee, calcaneus) (Fig. 3a–c). We were able to determine four stages according to the severity of the morphological alterations. Specific findings included epimetaphyseal osteolysis, followed by confluent microcallus formation and band-like sclerosis along the growth plates. These stress fractures were prone to eventually collapsing, leading to severe fractures and deformities in some cases (Supplementary Fig. 1).Fig. 3 Cone beam CT (CBCT) imaging. Differentiation and classification of different disease severities based on CBCT imaging a in the distal tibia, b around the knee (distal femur and proximal tibia) and c in the calcaneus To further characterize the skeletal changes on a microscopic level, a biopsy was obtained from the lateral (nonfractured) part of the tibial plateau. In this patient, the stress fracture of the medial tibial plateau was already visible on conventional radiography (Fig. 4a) and later confirmed by MRI (Fig. 4b). The tibial plateau was subsequently imaged by contact radiography (Fig. 4c). Histological quantification was performed in comparison with an age-matched control (Fig. 4d). The tibial bone microstructure of the patient with MTX osteopathy was characterized by an unchanged BV/TV and a higher trabecular number but a lower trabecular thickness (Fig. 4e–g). Furthermore, a low number of osteoblasts but a high number of osteoclasts were detected, leading to a lower osteoblast surface and a higher osteoclast surface compared to those of the control (Fig. 4h–j). Backscattered electron imaging confirmed highly prevalent eroded surfaces but an overall similar bone mineral density distribution (Fig. 4k, l). Additional histomorphometric analysis of three fracture biopsies from the distal tibiae and the calcaneus revealed the presence of fracture calluses and woven bone but no detection of osteonecrosis. This was associated with a generally high bone turnover (Fig. 5).Fig. 4 Biopsy studies (81-year-old woman) in the lateral (nonfractured) tibial plateau. a Anteroposterior radiograph showing the fracture of the medial tibial plateau (red arrow). b Coronal MRI, PD-weighted sequence. c Contact radiography (lateral view) of the resected tibial plateau, a anterior, p posterior. d Histological overview, von Kossa staining in the patient and the control. e–g Quantification of BV/TV, Tb.N and Tb.Th. h Visible osteoclasts on the surface of a trabecula, toluidine blue staining. i, j Osteoblast and osteoclast surface. k Image obtained by backscattered electron imaging with eroded surfaces (asterisks). l BMDD histograms Fig. 5 Biopsies from different fracture sites reveal interference with fracture healing with chronic callus formation and osteoidosis but an absence of osteonecrosis. a Representative histological images, toluidine blue staining. Left (no. 1): distal tibia (64-year-old woman) showing fracture callus; middle and right (nos. 2, 3): distal tibia and calcaneus (71-year-old woman) with woven bone formation. b Histomorphometric quantification in the individual biopsies nos. 1–3 including osteoid volume per bone volume (OV/BV), osteoblast surface per bone surface (Ob.S/BS) and osteoclast surface per bone surface (Oc.S/BS) Treatment Outcome and Follow-Up MTX treatment was discontinued or replaced in 27/34 patients, while the remaining seven patients underwent MTX dose reduction. The discontinuation of MTX, in consultation with an expert rheumatologist, was well tolerated. MTX was replaced by azathioprine, low-dose glucocorticoids or monoclonal antibodies (e.g., IL-17). Moreover, a bone-specific therapy consisting of denosumab, teriparatide or combined denosumab–teriparatide was initiated on the basis of the risk profiles and pre-existing comorbidities. Follow-up examinations in ten patients (8/10 MTX discontinuation) revealed that no further stress fractures had occurred, while treatment modifications were well tolerated and overall mobility was improved. Most patients showed an adequate increase or at least stable BMD values in the lumbar spine and hip (Table 1). Five of ten patients reported major improvements regarding mobility and pain levels, while the other five patients reported minor improvements (Fig. 6a). Of the 5/10 patients with a major improvement, all had discontinued their MTX medication. The two patients who continued MTX had only minor clinical improvements and showed no increase in BMD. Although MRI follow-up could not be performed in all patients, healing of stress fractures was observed in individual patients who underwent follow-up MRI (Fig. 6b). Successful remobilization and return to daily activities were achieved in all cases. Timed Up and Go testing confirmed the regained mobility, although the reference of 10 s was not reached in most patients (Fig. 6c). The CRT indicated slow but possible rises from a chair, while both tests were not possible in any of the patients at the initial presentation (Fig. 6d). The analysis of bone microarchitecture in the tibia using HR-pQCT revealed constant trabecular parameters and marked improvements in cortical parameters at follow-up (Fig. 6e–j, Supplementary Tab. 1 + 2).Table 1 BMD and corresponding T-scores assessed by DXA in the lumbar spine (LS) and hip of ten patients with follow-up measurements Pat. Time interval (yr.) MTX Specific therapy CC BMD LS T-score LS BMD hip T-score hip Initial Follow-up Change (%) Initial Follow-up Change (SD) Initial Follow -up Change (%) Initial Follow -up Change (SD) 1 1.5 DIS DATA ++ 1.027 1.012 − 1.5 − 1.3 − 1.3 ± 0.0 0.810 0.831 + 2.6 − 1.4 − 1.2 + 0.2 2 2.3 CON DATA + n/a n/a n/a n/a n/a n/a 0.671 0.660 − 1.6 − 3.2 − 3.6 − 0.4 3 4.0 DIS DATA + 0.869 0.957 + 10.1 − 2.8 − 2.0 + 0.8 0.675 0.686 + 1.6 − 2.7 − 2.6 + 0.1 4 4.0 DIS DATA ++ 1.121 1.054 − 6.0 − 0.8 − 0.9 − 0.1 0.689 0.717 + 4.1 − 2.6 − 2.2 + 0.4 5 3.8 DIS DATA ++ 0.709 0.788 + 11.1 − 3.8 − 2.4 + 1.4 0.717 0.726 + 1.3 − 2.4 0.1 + 2.5 6 1.6 DIS no ++ 0.713 0.764 + 7.2 − 4.0 − 3.6 + 0.4 0.609 0.580 − 4.8 − 3.3 − 3.5 − 0.2 7 1.0 DIS DATA ++ 0.950 0.962 + 1.3 − 1.8 − 1.7 + 0.1 0.784 0.797 + 1.7 − 1.6 − 1.5 + 0.1 8 1.6 DIS no + 0.743 0.785 + 5.7 − 3.5 − 3.3 + 0.2 0.660 0.689 + 4.4 − 2.7 − 2.4 + 0.3 9 1.0 DIS DATA + 1.028 1.078 + 4.9 − 1.1 − 0.7 + 0.4 0.779 0.864 + 10.9 − 1.8 − 1.1 + 0.7 10 5.0 CON TPT → D’mab + 0.988 0.961 − 2.7 − 1.8 − 1.8 ± 0.0 n/a n/a n/a n/a n/a n/a Changes are presented as percent (%) or standard deviation (SD) Yr. years, DIS discontinued, CON continued, DATA denosumab and teriparatide, D’mab denosumab, TPT teriparatide, CC clinical course (++/+ major/minor clinical improvement), n/a not available due to previous orthopedic surgery such as spondylodesis or hip replacement Fig. 6 Follow-up and treatment response. a Subjective clinical course (++ major improvement, + minor improvement, − no improvement in mobility and pain). b MRI, T1-weighted, sagittal sequence. c Individual results for the Timed Up and Go test at follow-up. d Chair rising test (CRT). e–j HR-pQCT at initial presentation and follow-up (distal tibia for all panels). e Trabecular BMD. f Trabecular number. g Cortical BMD. h Cortical thickness. i HR-pQCT image at 0 and 14 months indicating increasing cortical thickness (white arrows). j HR-pQCT changes converted to %-change/1 year Discussion While stress fractures in patients with long-term MTX use have been repeatedly described, detailed knowledge on the underlying skeletal alterations in a larger patient collective was not available to date. From a clinical perspective, our results suggest that long-term MTX treatment may have negative effects on bone metabolism and quality in certain patient groups, which was expressed by stress fractures with a unique band- or meander-shaped appearance along the growth plate. Indeed, our MRI-based morphological characterization therefore corresponds well to the first reports on MTX osteopathy [3, 4, 6]. In other words, this comprehensive imaging analysis illustrates that the detected uniquely shaped stress fractures constitute the hallmark of MTX osteopathy. Even in different rheumatic diseases, this typical clinical and radiological picture was detected, emphasizing the concept of significant MTX side effects rather than concomitant phenomena of the underlying diseases. At the same time, stress fractures due to other reasons including overuse, osteomalacia (e.g., calcium malabsorption, phosphate wasting) or inflammatory conditions are typically not characterized by a meander-shaped morphology along the growth plates. The skeletal alterations were subsequently characterized by CBCT, where a consistent radiographic pattern of epimetaphyseal osteolysis and band-like sclerosis (i.e., microcallus) was found. In association with this pathognomonic imaging appearance detected by MRI and CBCT, we observed a consistent bone microstructure and turnover pattern. Namely, the skeletal deterioration was further characterized by HR-pQCT, where pronounced trabecular thinning was detected. Bone turnover was often characterized by a combination of high bone-specific alkaline phosphatase levels, low osteocalcin levels indicating low bone formation, and elevated bone resorption parameters (i.e., urinary DPD cross links). This dissociation between low bone formation and high bone resorption was recapitulated in a bone biopsy obtained from a patient with MTX osteopathy, while three biopsies from fracture sites indicated interference with fracture healing but an absence of osteonecrosis. In sum, the detected bone turnover dissociation is most likely a major contributing factor for the detected long-term skeletal complications. Based on our findings, we suggest that any symptomatic patient with lower extremity pain, loss of mobility and long-term MTX treatment should be rigorously screened for stress fractures and skeletal status (i.e., osteoporosis) using MRI, DXA and laboratory analyses. CBCT represents a valuable tool to further image these osseous alterations at a high resolution while simultaneously reducing radiation exposure [22]. While the correct diagnosis can help to ensure the best medical care in terms of appropriate treatment, there is a risk that stress fractures will progress towards extensive bone and joint destruction (i.e., collapse) if not treated properly and MTX is continued in these patients (Supplementary Fig. 1). Although we do understand that a discontinuation of MTX might not be feasible in all patients with different rheumatic inflammatory diseases, MTX discontinuation should always be considered in patients with MTX osteopathy. As the diagnosed stress fractures were accompanied by osteoporosis or osteopenia, according to DXA, and a dissociation of low bone formation and elevated bone resorption in most cases, we treated most our patients with both denosumab (anti-resorptive) and teriparatide (osteoanabolic) according to the previously published DATA study [32]. We additionally guaranteed sufficient 25-OH-D3 levels, as this was found to be beneficial for reducing the risk of stress fractures [33]. In combination with a discontinuation of MTX treatment, the analyzed subset of ten patients recovered from stress fractures and regained their mobility, which was associated with increased BMD levels and improved cortical microstructure in most patients. Close rheumatologic monitoring was carried out to avoid flares of the underlying autoimmune disease. The denosumab–teriparatide combination therapy was previously shown to lead to the highest increases in BMD [32] before the sclerostin antibody romosozumab entered clinical research [34]. However, it is currently not known whether these findings can be applied to special patient groups such as RA patients with MTX osteopathy. In rats with MTX-induced osteopathy, anti-resorptive treatment was found to be effective [11]. The addition of denosumab to MTX has also been found to be an effective treatment option in the prevention of further joint destruction in patients with RA [35]. We are aware that our study only offers correlative evidence regarding MTX osteopathy as a clinical entity that is described by pathognomonic and nonself-limiting stress fractures. Our results do not provide sufficient information in terms of causality and stress fracture prevalence due to long-term MTX treatment, and future studies will have to elaborate on the epidemiologic aspects of this burden. Our data also do not allow us to determine whether MTX discontinuation or the initiated bone-specific therapy is more effective regarding the achieved clinical improvements. It is interesting to note that from the ten patients analyzed at follow-up, the two patients who continued MTX showed minor clinical improvements and no increase in BMD. Finally, other conditions, such as inflammation, corticosteroid use or postmenopausal status, also promote an impairment in bone health. Nonetheless, several studies have linked MTX to deteriorated bone quality. Reduced BMD was observed with MTX use compared to no MTX use among patients with RA [36]. In a prospective trial of 329 RA patients, a negative annual BMD change was significantly associated with MTX dosage [37]. Moreover, even in several other reported cases of patients with RA and stress fractures, most of them had previously undergone long-term MTX therapy [38–40]. On the other hand, MTX was associated with neither improved nor worsened bone microstructure in patients with PsA, while DMARD use (such as TNFalpha inhibitors or IL-17A antibodies) led to improved microstructure [41]. In general, it remains a major issue to develop optimal drug strategies for treating bone problems in patients with inflammatory diseases [42]. Taken together, we have provided clinical evidence of the potential negative effects of long-term MTX use on bone quality, associated with peculiar stress fractures that can be positively influenced by a discontinuation of MTX together with the initiation of a combined denosumab and teriparatide treatment. Whether the observed improvements in stress fractures and changes in quality translate into reductions in stress fracture risk over the long term remains to be evaluated in larger series. Electronic supplementary material Below is the link to the electronic supplementary material.Supplementary file1 (PDF 1308 kb) Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Tim Rolvien and Nico Maximilian Jandl contributed equally to this work. Acknowledgements The authors thank Andrea Thieke, Olga Winter and Elke Leicht for technical assistance with the histological sections. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with Ethical Standards Conflict of interest Tim Rolvien, Nico Maximilian Jandl, Julian Stürznickel, Frank Timo Beil, Ina Kötter, Ralf Oheim, Ansgar W Lohse, Florian Barvencik and Michael Amling declare that they have no conflict of interest. Human and Animal Rights All procedures are in accordance with the ethical standards as laid down in the 1964 Declaration of Helsinki and its later amendments or comparable ethical standards. Informed Consent Informed consent was obtained from all individual patients included in this study.
METHOTREXATE
DrugsGivenReaction
CC BY
33064170
18,451,574
2021-02
What was the dosage of drug 'METHOTREXATE'?
Clinical and Radiological Characterization of Patients with Immobilizing and Progressive Stress Fractures in Methotrexate Osteopathy. Methotrexate (MTX) is one of the most commonly prescribed drugs for autoimmune rheumatic diseases. As there is no consensus on its negative effects on bone, the purpose of this investigation was to determine the clinical spectrum of patients with stress fractures due to long-term MTX treatment (i.e., MTX osteopathy). We have retrospectively analyzed data from 34 patients with MTX treatment, severe lower extremity pain and immobilization. MRI scans, bone turnover markers, bone mineral density (DXA) and bone microarchitecture (HR-pQCT) were evaluated. Stress fractures were also imaged with cone beam CT. While the time between clinical onset and diagnosis was prolonged (17.4 ± 8.6 months), the stress fractures had a pathognomonic appearance (i.e., band-/meander-shaped, along the growth plate) and were diagnosed in the distal tibia (53%), the calcaneus (53%), around the knee (62%) and at multiple sites (68%). Skeletal deterioration was expressed by osteoporosis (62%) along with dissociation of low bone formation and increased bone resorption. MTX treatment was discontinued in 27/34 patients, and a combined denosumab-teriparatide treatment initiated. Ten patients re-evaluated at follow-up (2.6 ± 1.5 years) had improved clinically in terms of successful remobilization. Taken together, our findings provide the first in-depth skeletal characterization of patients with pathognomonic stress fractures after long-term MTX treatment. Introduction Methotrexate (MTX) is a folate antagonist that is used in low doses (5–25 mg/week) in the first-line treatment of rheumatoid arthritis (RA), as well as in other inflammatory diseases such as systemic lupus erythematosus (SLE). It acts by inhibiting dihydrofolate reductase, which is an essential factor in DNA and RNA synthesis, but also has far-reaching anti-inflammatory and immunoregulatory effects [1, 2]. While the effects of MTX on bone metabolism have not been completely elucidated, there is both clinical and in vitro evidence for its adverse effects on bone. MTX osteopathy was first described in 1984 in children who had undergone prolonged maintenance therapy with oral MTX due to acute lymphocytic leukemia (ALL) [3], where distal femoral and tibial fractures with thick dense provisional zones of calcification were detected. In the 1990s, several clinical reports described “MTX osteopathy”, including low bone mass and stress fractures of the distal and proximal tibiae [4–6]. Since then, stress fractures of the proximal tibia have also been described in other patients after long-term MTX use [7, 8]. Inhibitory effects of MTX on osteoblastic bone formation have been detected in human biopsies as well as in murine bone cells [4, 9, 10], and bone mechanical properties were impaired in MTX-treated rats [11]. Although the documented effects of MTX include stimulation of both pro-inflammatory and anti-inflammatory pathways, increased cytokine production may be a possible mechanism for tissue damage in certain conditions such as in MTX osteopathy. In this regard, MTX treatment in cell culture has been linked to a dose-dependent increase in pro-inflammatory cytokines such as IL-1 and IL-6 [12]. At higher doses, MTX use led to increased TNF-α levels and promoted osteoclastogenesis [13]. MTX osteopathy has also been questioned based on the observation that MTX treatment in small patient cohorts and after short-term follow-up did not result in changes in bone turnover or bone density [14], and comparable bone loss compared to other disease-modifying antirheumatic drugs (DMARDs) [15]. Therefore, the clinical relevance of MTX osteopathy remains unclear. In general, the impairment of bone mineral density and quality as well as the increased risk of fracture have been reported in a variety of rheumatic diseases; however, they are often attributed to inflammatory processes and corticosteroid therapy [16–18]. As there are indications of low bone formation and elevated bone resorption in MTX osteopathy, a combination of anti-resorptive (i.e., denosumab) and osteoanabolic (i.e., teriparatide) therapy might be the most logical treatment option to improve bone strength and recover from stress fractures. We have previously demonstrated recovery from stress fractures in an SLE patient with MTX osteopathy after MTX discontinuation and combined denosumab–teriparatide treatment [19]. Here, we present a clinical characterization of 34 patients suffering from stress fractures after long-term methotrexate use for different underlying rheumatologic diseases and outline their positive response to denosumab and teriparatide treatment in a subset of ten patients. Methods Subjects Thirty-four patients with long-term (> 3 years) methotrexate use and stress fractures were included in this study. All patient data were evaluated in a retrospective and anonymized design. Data were analyzed according to the rules of the local ethics committee of the University Medical Center Hamburg-Eppendorf, Germany. Skeletal Assessment and Imaging Studies While magnetic resonance imaging (MRI) was performed for individual stress fracture detection, we also determined the areal bone mineral density (aBMD) using dual-energy X-ray absorptiometry (Lunar iDXA; GE Healthcare; Madison, WI, USA) and bone turnover markers from serum and urine samples in all patients. The measured serum markers included 25-hydroxyvitamin D (25-OH-D3), parathyroid hormone (PTH), osteocalcin (Oc), bone-specific alkaline phosphatase (BAP), and deoxypyridinoline cross-links in the urine (DPD). Furthermore, bone microarchitecture was analyzed in 30/34 patients using high-resolution peripheral quantitative computed tomography (HR-pQCT; XtremeCT, Scanco Medical, Switzerland). In the remaining four patients, HR-pQCT could not be performed due to bilateral distal tibia fractures. Scans were performed in the nonfractured distal tibia following a standardized procedure using the standard in vivo patient evaluation protocol [20, 21]. Specifically, we analyzed the trabecular bone mineral density (Tb.BMD), trabecular number (Tb.N), trabecular thickness (Tb.Th) and cortical bone mineral density (Ct.BMD). Stress fractures were also imaged in ten patients using cone beam computed tomography (CBCT) working at 90 kV, 40 mAs with a field of view of 16 × 16 ×  13 cm and slice thickness of 0.2 mm (SCS MedSeries H22, Planmed Oy, Helsinki, Finland). CBCT is a novel imaging technique using divergent X-rays that form a cone, which is increasingly used in extremity imaging, though its initial application was in dentistry and maxillofacial surgery [22, 23]. CBCT enables better fracture detection at extremity sites compared to standard radiography [24]. Furthermore, the spatial resolution of CBCT is higher and the radiation dose exposure is lower compared to conventional multislice CT [25]. Bone Biopsy Studies We obtained four bone biopsies from individuals with long-term MTX treatment and stress fractures. The first biopsy was obtained from an 81-year-old woman suffering from a stress fracture of the proximal right tibia. The biopsy was obtained from the lateral (unaffected) part of the tibial plateau in the course of total knee arthroplasty (TKA) and compared to a biopsy obtained from a 79-year-old female patient with primary osteoarthritis undergoing TKA. The other three biopsies were obtained from fracture sites as a part of a diagnostic work-up in these patients. While the second biopsy was obtained from the distal tibia of a 64-year-old patient previously reported elsewhere [19], the third and fourth biopsies were taken from the distal tibia and the calcaneus of a 71-year-old woman with RA. The specimens were fixed in 3.7% formaldehyde, dehydrated, embedded in methyl-methacrylate, and cut on a Microtec rotation microtome (CVT 4060E, Micro Tec, Walldorf, Germany). Afterwards the 5-µm sections were stained with toluidine blue and von Kossa. Histomorphometric analysis was performed according to the ASBMR nomenclature committee [26]. BV/TV, Tb.N, and Tb.Th as well as osteoid volume per bone volume (OV/BV) were evaluated in the von Kossa-stained sections. The osteoblast surface per bone surface (Ob.S/BS) and the osteoclast surface per bone surface (Oc.S/BS) were evaluated from the toluidine blue-stained sections. Quantitative backscattered electron imaging (qBEI) was performed on the first biopsy using a scanning electron microscope (LEO 435 VP; LEO Electron Microscopy Ltd., Cambridge, England) with a backscattered electron detector (Type 202; K.E. Developments Ltd., Cambridge, England), as described previously [27]. The scanning electron microscope was operated at 20 kV and 680 pA at a constant working distance. The acquired images were analyzed using a customized MATLAB (The MathWorks, Inc., Natick, Massachusetts, USA) script. QBEI was used to measure the bone mineral density distribution based on the generated gray values that represent the mean calcium content (mean Ca-Wt%). For both samples, calcium distribution curves were calculated. Treatment Intervention and Follow-Up Ten patients were evaluated at a follow-up visit at 2.6 ± 1.5 years (min. 1 year, max. 5 years). MTX treatment had been discontinued in 8/10 patients in consultation with a rheumatologist, while the MTX dose was reduced in the remaining 2 patients. Seven out of 10 patients were treated with a subcutaneous administration of denosumab 60 mg (Prolia®, Amgen, USA) every 6 months in combination with a daily subcutaneous administration of teriparatide 20 µg (Forsteo®, Eli Lilly, USA). While two patients were not treated with this bone-specific therapy due to individual contraindications, one patient was treated with teriparatide only. The two patients with no bone-specific treatment had discontinued their MTX medication. All patients received 20,000 IE vitamin D3 and 1 g of dietary calcium daily. At follow-up, clinical reexamination and an HR-pQCT scan were performed. The clinical course was evaluated, and patients were asked about subjective changes in mobility and pain (++/+ major/minor improvement, −/− no improvement/worsened). Furthermore, we performed the Timed Up and Go test and compared the achieved time to previously published reference values [28]. The chair rising test (CRT) was performed as described previously [29], and the results were compared to reference data [30]. Statistical Analysis All data were evaluated using GraphPad Prism® (GraphPad Software, La Jolla, CA, USA). Data are presented as scatter plots with additional labeling of the mean value ± the standard deviation (SD). After checking for normal distribution, the paired t-test was used to compare measurement results from the initial presentation and follow-up. Furthermore, the percent change per year was calculated. P-values of 0.05 or less were considered statistically significant. Results Clinical Characterization A total of 34 patients were evaluated, of whom all had load-dependent pain of the lower limb without adequate trauma in the past. MRI pointed to band- or meander-shaped stress fractures paralleling the former provisional zones of calcification and growth plates in all cases. The most frequent location was the distal tibia (53%) and the calcaneus (53%), followed by the proximal tibia (44%) and the distal femur (18%). In 68% of the patients, we observed stress fractures at multiple and/or bilateral locations (Fig. 1a–e). No association between the time interval of clinical onset and diagnosis and the MTX treatment durations or doses could be detected.Fig. 1 MRI morphology of stress fractures in MTX osteopathy. Band-like stress fractures in proton density (PD)-weighted fat-suppressed (FS) turbo spin echo (TSE) MRI sequences are seen. a Distal tibia, coronal and sagittal plane of two different patients. b Calcaneus, coronal and sagittal plane of two different patients. c Proximal tibia/distal femur, coronal plane. d Multiple stress fractures were found in 68% of the patients. e Bar graph indicating the regional distribution of stress fractures in n = 34 patients (frequency in % for each skeletal site) MTX osteopathy was diagnosed mostly in females (88%, Fig. 2a). The MTX dosage was 18.6 ± 4.9 mg (15–25 mg) weekly. Importantly, a prolonged time between clinical onset and diagnosis of 17.4 ± 8.6 months (5–36 months) was noted. Most patients had a history of corticosteroid treatment; however, upon presentation at our clinic, only 12/34 patients (35.3%) received low-dose oral prednisone (< 5 mg) (Fig. 2b). MTX treatment had been prescribed due to RA (26/34 patients), psoriatic arthritis (4/34 patients), SLE (2/34 patients), polymyalgia rheumatica (1/34 patients) or ankylosing spondylitis (1/34 patients) (Fig. 1c). Only two patients had vertebral fractures. We observed a peak in the occurrence of MTX osteopathy at the age of 70–79 years (Fig. 2d).Fig. 2 Clinical characteristics and bone mineral density, turnover and microstructure. a Eighty-eight percent of the affected patients were female. b Prednisone/no prednisone treatment. c Distribution of the different rheumatic diseases. d Age distribution. e, f DXA T-score at the lumbar spine (LS) and total hip. g, h Serum bone-specific alkaline phosphatase (BAP) and osteocalcin levels (both bone formation) and i urinary deoxypyridinoline (DPD) (bone resorption). Gray boxes indicate reference ranges. j Trabecular number (Tb. N), k trabecular thickness (Tb. Th) and l cortical BMD compared to age- and sex-matched reference data [31] A total of 21/34 (61.8%) of the patients were diagnosed with osteoporosis (i.e., BMD T-score ≤ − 2.5), and the remaining patients were diagnosed with osteopenia, according to the DXA measurements (Fig. 2e, f). Biochemical bone turnover analyses revealed higher levels in the bone-specific alkaline phosphatase than in the osteocalcin levels compared to reference values (Fig. 2g, h). In 18/32 patients (56%), we detected elevated bone resorption markers in the urine (i.e., DPD cross links) (Fig. 2i). HR-pQCT analysis revealed an almost normal trabecular number in most patients and a more severe reduction of trabecular thickness and cortical BMD in the distal tibia compared to reference values (Fig. 2j–l). While two patients presented with 25-hydroxy-vitamin D3 levels < 20 µg/l, ten patients had 25-OH-D3 levels < 30 µg/l. All patients received oral vitamin D supplementation. Cone Beam CT (CBCT) Imaging and Biopsy Findings CBCT imaging demonstrated osseous alterations around the typical stress fracture localizations (distal tibia, around the knee, calcaneus) (Fig. 3a–c). We were able to determine four stages according to the severity of the morphological alterations. Specific findings included epimetaphyseal osteolysis, followed by confluent microcallus formation and band-like sclerosis along the growth plates. These stress fractures were prone to eventually collapsing, leading to severe fractures and deformities in some cases (Supplementary Fig. 1).Fig. 3 Cone beam CT (CBCT) imaging. Differentiation and classification of different disease severities based on CBCT imaging a in the distal tibia, b around the knee (distal femur and proximal tibia) and c in the calcaneus To further characterize the skeletal changes on a microscopic level, a biopsy was obtained from the lateral (nonfractured) part of the tibial plateau. In this patient, the stress fracture of the medial tibial plateau was already visible on conventional radiography (Fig. 4a) and later confirmed by MRI (Fig. 4b). The tibial plateau was subsequently imaged by contact radiography (Fig. 4c). Histological quantification was performed in comparison with an age-matched control (Fig. 4d). The tibial bone microstructure of the patient with MTX osteopathy was characterized by an unchanged BV/TV and a higher trabecular number but a lower trabecular thickness (Fig. 4e–g). Furthermore, a low number of osteoblasts but a high number of osteoclasts were detected, leading to a lower osteoblast surface and a higher osteoclast surface compared to those of the control (Fig. 4h–j). Backscattered electron imaging confirmed highly prevalent eroded surfaces but an overall similar bone mineral density distribution (Fig. 4k, l). Additional histomorphometric analysis of three fracture biopsies from the distal tibiae and the calcaneus revealed the presence of fracture calluses and woven bone but no detection of osteonecrosis. This was associated with a generally high bone turnover (Fig. 5).Fig. 4 Biopsy studies (81-year-old woman) in the lateral (nonfractured) tibial plateau. a Anteroposterior radiograph showing the fracture of the medial tibial plateau (red arrow). b Coronal MRI, PD-weighted sequence. c Contact radiography (lateral view) of the resected tibial plateau, a anterior, p posterior. d Histological overview, von Kossa staining in the patient and the control. e–g Quantification of BV/TV, Tb.N and Tb.Th. h Visible osteoclasts on the surface of a trabecula, toluidine blue staining. i, j Osteoblast and osteoclast surface. k Image obtained by backscattered electron imaging with eroded surfaces (asterisks). l BMDD histograms Fig. 5 Biopsies from different fracture sites reveal interference with fracture healing with chronic callus formation and osteoidosis but an absence of osteonecrosis. a Representative histological images, toluidine blue staining. Left (no. 1): distal tibia (64-year-old woman) showing fracture callus; middle and right (nos. 2, 3): distal tibia and calcaneus (71-year-old woman) with woven bone formation. b Histomorphometric quantification in the individual biopsies nos. 1–3 including osteoid volume per bone volume (OV/BV), osteoblast surface per bone surface (Ob.S/BS) and osteoclast surface per bone surface (Oc.S/BS) Treatment Outcome and Follow-Up MTX treatment was discontinued or replaced in 27/34 patients, while the remaining seven patients underwent MTX dose reduction. The discontinuation of MTX, in consultation with an expert rheumatologist, was well tolerated. MTX was replaced by azathioprine, low-dose glucocorticoids or monoclonal antibodies (e.g., IL-17). Moreover, a bone-specific therapy consisting of denosumab, teriparatide or combined denosumab–teriparatide was initiated on the basis of the risk profiles and pre-existing comorbidities. Follow-up examinations in ten patients (8/10 MTX discontinuation) revealed that no further stress fractures had occurred, while treatment modifications were well tolerated and overall mobility was improved. Most patients showed an adequate increase or at least stable BMD values in the lumbar spine and hip (Table 1). Five of ten patients reported major improvements regarding mobility and pain levels, while the other five patients reported minor improvements (Fig. 6a). Of the 5/10 patients with a major improvement, all had discontinued their MTX medication. The two patients who continued MTX had only minor clinical improvements and showed no increase in BMD. Although MRI follow-up could not be performed in all patients, healing of stress fractures was observed in individual patients who underwent follow-up MRI (Fig. 6b). Successful remobilization and return to daily activities were achieved in all cases. Timed Up and Go testing confirmed the regained mobility, although the reference of 10 s was not reached in most patients (Fig. 6c). The CRT indicated slow but possible rises from a chair, while both tests were not possible in any of the patients at the initial presentation (Fig. 6d). The analysis of bone microarchitecture in the tibia using HR-pQCT revealed constant trabecular parameters and marked improvements in cortical parameters at follow-up (Fig. 6e–j, Supplementary Tab. 1 + 2).Table 1 BMD and corresponding T-scores assessed by DXA in the lumbar spine (LS) and hip of ten patients with follow-up measurements Pat. Time interval (yr.) MTX Specific therapy CC BMD LS T-score LS BMD hip T-score hip Initial Follow-up Change (%) Initial Follow-up Change (SD) Initial Follow -up Change (%) Initial Follow -up Change (SD) 1 1.5 DIS DATA ++ 1.027 1.012 − 1.5 − 1.3 − 1.3 ± 0.0 0.810 0.831 + 2.6 − 1.4 − 1.2 + 0.2 2 2.3 CON DATA + n/a n/a n/a n/a n/a n/a 0.671 0.660 − 1.6 − 3.2 − 3.6 − 0.4 3 4.0 DIS DATA + 0.869 0.957 + 10.1 − 2.8 − 2.0 + 0.8 0.675 0.686 + 1.6 − 2.7 − 2.6 + 0.1 4 4.0 DIS DATA ++ 1.121 1.054 − 6.0 − 0.8 − 0.9 − 0.1 0.689 0.717 + 4.1 − 2.6 − 2.2 + 0.4 5 3.8 DIS DATA ++ 0.709 0.788 + 11.1 − 3.8 − 2.4 + 1.4 0.717 0.726 + 1.3 − 2.4 0.1 + 2.5 6 1.6 DIS no ++ 0.713 0.764 + 7.2 − 4.0 − 3.6 + 0.4 0.609 0.580 − 4.8 − 3.3 − 3.5 − 0.2 7 1.0 DIS DATA ++ 0.950 0.962 + 1.3 − 1.8 − 1.7 + 0.1 0.784 0.797 + 1.7 − 1.6 − 1.5 + 0.1 8 1.6 DIS no + 0.743 0.785 + 5.7 − 3.5 − 3.3 + 0.2 0.660 0.689 + 4.4 − 2.7 − 2.4 + 0.3 9 1.0 DIS DATA + 1.028 1.078 + 4.9 − 1.1 − 0.7 + 0.4 0.779 0.864 + 10.9 − 1.8 − 1.1 + 0.7 10 5.0 CON TPT → D’mab + 0.988 0.961 − 2.7 − 1.8 − 1.8 ± 0.0 n/a n/a n/a n/a n/a n/a Changes are presented as percent (%) or standard deviation (SD) Yr. years, DIS discontinued, CON continued, DATA denosumab and teriparatide, D’mab denosumab, TPT teriparatide, CC clinical course (++/+ major/minor clinical improvement), n/a not available due to previous orthopedic surgery such as spondylodesis or hip replacement Fig. 6 Follow-up and treatment response. a Subjective clinical course (++ major improvement, + minor improvement, − no improvement in mobility and pain). b MRI, T1-weighted, sagittal sequence. c Individual results for the Timed Up and Go test at follow-up. d Chair rising test (CRT). e–j HR-pQCT at initial presentation and follow-up (distal tibia for all panels). e Trabecular BMD. f Trabecular number. g Cortical BMD. h Cortical thickness. i HR-pQCT image at 0 and 14 months indicating increasing cortical thickness (white arrows). j HR-pQCT changes converted to %-change/1 year Discussion While stress fractures in patients with long-term MTX use have been repeatedly described, detailed knowledge on the underlying skeletal alterations in a larger patient collective was not available to date. From a clinical perspective, our results suggest that long-term MTX treatment may have negative effects on bone metabolism and quality in certain patient groups, which was expressed by stress fractures with a unique band- or meander-shaped appearance along the growth plate. Indeed, our MRI-based morphological characterization therefore corresponds well to the first reports on MTX osteopathy [3, 4, 6]. In other words, this comprehensive imaging analysis illustrates that the detected uniquely shaped stress fractures constitute the hallmark of MTX osteopathy. Even in different rheumatic diseases, this typical clinical and radiological picture was detected, emphasizing the concept of significant MTX side effects rather than concomitant phenomena of the underlying diseases. At the same time, stress fractures due to other reasons including overuse, osteomalacia (e.g., calcium malabsorption, phosphate wasting) or inflammatory conditions are typically not characterized by a meander-shaped morphology along the growth plates. The skeletal alterations were subsequently characterized by CBCT, where a consistent radiographic pattern of epimetaphyseal osteolysis and band-like sclerosis (i.e., microcallus) was found. In association with this pathognomonic imaging appearance detected by MRI and CBCT, we observed a consistent bone microstructure and turnover pattern. Namely, the skeletal deterioration was further characterized by HR-pQCT, where pronounced trabecular thinning was detected. Bone turnover was often characterized by a combination of high bone-specific alkaline phosphatase levels, low osteocalcin levels indicating low bone formation, and elevated bone resorption parameters (i.e., urinary DPD cross links). This dissociation between low bone formation and high bone resorption was recapitulated in a bone biopsy obtained from a patient with MTX osteopathy, while three biopsies from fracture sites indicated interference with fracture healing but an absence of osteonecrosis. In sum, the detected bone turnover dissociation is most likely a major contributing factor for the detected long-term skeletal complications. Based on our findings, we suggest that any symptomatic patient with lower extremity pain, loss of mobility and long-term MTX treatment should be rigorously screened for stress fractures and skeletal status (i.e., osteoporosis) using MRI, DXA and laboratory analyses. CBCT represents a valuable tool to further image these osseous alterations at a high resolution while simultaneously reducing radiation exposure [22]. While the correct diagnosis can help to ensure the best medical care in terms of appropriate treatment, there is a risk that stress fractures will progress towards extensive bone and joint destruction (i.e., collapse) if not treated properly and MTX is continued in these patients (Supplementary Fig. 1). Although we do understand that a discontinuation of MTX might not be feasible in all patients with different rheumatic inflammatory diseases, MTX discontinuation should always be considered in patients with MTX osteopathy. As the diagnosed stress fractures were accompanied by osteoporosis or osteopenia, according to DXA, and a dissociation of low bone formation and elevated bone resorption in most cases, we treated most our patients with both denosumab (anti-resorptive) and teriparatide (osteoanabolic) according to the previously published DATA study [32]. We additionally guaranteed sufficient 25-OH-D3 levels, as this was found to be beneficial for reducing the risk of stress fractures [33]. In combination with a discontinuation of MTX treatment, the analyzed subset of ten patients recovered from stress fractures and regained their mobility, which was associated with increased BMD levels and improved cortical microstructure in most patients. Close rheumatologic monitoring was carried out to avoid flares of the underlying autoimmune disease. The denosumab–teriparatide combination therapy was previously shown to lead to the highest increases in BMD [32] before the sclerostin antibody romosozumab entered clinical research [34]. However, it is currently not known whether these findings can be applied to special patient groups such as RA patients with MTX osteopathy. In rats with MTX-induced osteopathy, anti-resorptive treatment was found to be effective [11]. The addition of denosumab to MTX has also been found to be an effective treatment option in the prevention of further joint destruction in patients with RA [35]. We are aware that our study only offers correlative evidence regarding MTX osteopathy as a clinical entity that is described by pathognomonic and nonself-limiting stress fractures. Our results do not provide sufficient information in terms of causality and stress fracture prevalence due to long-term MTX treatment, and future studies will have to elaborate on the epidemiologic aspects of this burden. Our data also do not allow us to determine whether MTX discontinuation or the initiated bone-specific therapy is more effective regarding the achieved clinical improvements. It is interesting to note that from the ten patients analyzed at follow-up, the two patients who continued MTX showed minor clinical improvements and no increase in BMD. Finally, other conditions, such as inflammation, corticosteroid use or postmenopausal status, also promote an impairment in bone health. Nonetheless, several studies have linked MTX to deteriorated bone quality. Reduced BMD was observed with MTX use compared to no MTX use among patients with RA [36]. In a prospective trial of 329 RA patients, a negative annual BMD change was significantly associated with MTX dosage [37]. Moreover, even in several other reported cases of patients with RA and stress fractures, most of them had previously undergone long-term MTX therapy [38–40]. On the other hand, MTX was associated with neither improved nor worsened bone microstructure in patients with PsA, while DMARD use (such as TNFalpha inhibitors or IL-17A antibodies) led to improved microstructure [41]. In general, it remains a major issue to develop optimal drug strategies for treating bone problems in patients with inflammatory diseases [42]. Taken together, we have provided clinical evidence of the potential negative effects of long-term MTX use on bone quality, associated with peculiar stress fractures that can be positively influenced by a discontinuation of MTX together with the initiation of a combined denosumab and teriparatide treatment. Whether the observed improvements in stress fractures and changes in quality translate into reductions in stress fracture risk over the long term remains to be evaluated in larger series. Electronic supplementary material Below is the link to the electronic supplementary material.Supplementary file1 (PDF 1308 kb) Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Tim Rolvien and Nico Maximilian Jandl contributed equally to this work. Acknowledgements The authors thank Andrea Thieke, Olga Winter and Elke Leicht for technical assistance with the histological sections. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with Ethical Standards Conflict of interest Tim Rolvien, Nico Maximilian Jandl, Julian Stürznickel, Frank Timo Beil, Ina Kötter, Ralf Oheim, Ansgar W Lohse, Florian Barvencik and Michael Amling declare that they have no conflict of interest. Human and Animal Rights All procedures are in accordance with the ethical standards as laid down in the 1964 Declaration of Helsinki and its later amendments or comparable ethical standards. Informed Consent Informed consent was obtained from all individual patients included in this study.
18.6 +/- 4.9 MG (15-25 MG) WEEKLY
DrugDosageText
CC BY
33064170
18,451,574
2021-02
What was the outcome of reaction 'Stress fracture'?
Clinical and Radiological Characterization of Patients with Immobilizing and Progressive Stress Fractures in Methotrexate Osteopathy. Methotrexate (MTX) is one of the most commonly prescribed drugs for autoimmune rheumatic diseases. As there is no consensus on its negative effects on bone, the purpose of this investigation was to determine the clinical spectrum of patients with stress fractures due to long-term MTX treatment (i.e., MTX osteopathy). We have retrospectively analyzed data from 34 patients with MTX treatment, severe lower extremity pain and immobilization. MRI scans, bone turnover markers, bone mineral density (DXA) and bone microarchitecture (HR-pQCT) were evaluated. Stress fractures were also imaged with cone beam CT. While the time between clinical onset and diagnosis was prolonged (17.4 ± 8.6 months), the stress fractures had a pathognomonic appearance (i.e., band-/meander-shaped, along the growth plate) and were diagnosed in the distal tibia (53%), the calcaneus (53%), around the knee (62%) and at multiple sites (68%). Skeletal deterioration was expressed by osteoporosis (62%) along with dissociation of low bone formation and increased bone resorption. MTX treatment was discontinued in 27/34 patients, and a combined denosumab-teriparatide treatment initiated. Ten patients re-evaluated at follow-up (2.6 ± 1.5 years) had improved clinically in terms of successful remobilization. Taken together, our findings provide the first in-depth skeletal characterization of patients with pathognomonic stress fractures after long-term MTX treatment. Introduction Methotrexate (MTX) is a folate antagonist that is used in low doses (5–25 mg/week) in the first-line treatment of rheumatoid arthritis (RA), as well as in other inflammatory diseases such as systemic lupus erythematosus (SLE). It acts by inhibiting dihydrofolate reductase, which is an essential factor in DNA and RNA synthesis, but also has far-reaching anti-inflammatory and immunoregulatory effects [1, 2]. While the effects of MTX on bone metabolism have not been completely elucidated, there is both clinical and in vitro evidence for its adverse effects on bone. MTX osteopathy was first described in 1984 in children who had undergone prolonged maintenance therapy with oral MTX due to acute lymphocytic leukemia (ALL) [3], where distal femoral and tibial fractures with thick dense provisional zones of calcification were detected. In the 1990s, several clinical reports described “MTX osteopathy”, including low bone mass and stress fractures of the distal and proximal tibiae [4–6]. Since then, stress fractures of the proximal tibia have also been described in other patients after long-term MTX use [7, 8]. Inhibitory effects of MTX on osteoblastic bone formation have been detected in human biopsies as well as in murine bone cells [4, 9, 10], and bone mechanical properties were impaired in MTX-treated rats [11]. Although the documented effects of MTX include stimulation of both pro-inflammatory and anti-inflammatory pathways, increased cytokine production may be a possible mechanism for tissue damage in certain conditions such as in MTX osteopathy. In this regard, MTX treatment in cell culture has been linked to a dose-dependent increase in pro-inflammatory cytokines such as IL-1 and IL-6 [12]. At higher doses, MTX use led to increased TNF-α levels and promoted osteoclastogenesis [13]. MTX osteopathy has also been questioned based on the observation that MTX treatment in small patient cohorts and after short-term follow-up did not result in changes in bone turnover or bone density [14], and comparable bone loss compared to other disease-modifying antirheumatic drugs (DMARDs) [15]. Therefore, the clinical relevance of MTX osteopathy remains unclear. In general, the impairment of bone mineral density and quality as well as the increased risk of fracture have been reported in a variety of rheumatic diseases; however, they are often attributed to inflammatory processes and corticosteroid therapy [16–18]. As there are indications of low bone formation and elevated bone resorption in MTX osteopathy, a combination of anti-resorptive (i.e., denosumab) and osteoanabolic (i.e., teriparatide) therapy might be the most logical treatment option to improve bone strength and recover from stress fractures. We have previously demonstrated recovery from stress fractures in an SLE patient with MTX osteopathy after MTX discontinuation and combined denosumab–teriparatide treatment [19]. Here, we present a clinical characterization of 34 patients suffering from stress fractures after long-term methotrexate use for different underlying rheumatologic diseases and outline their positive response to denosumab and teriparatide treatment in a subset of ten patients. Methods Subjects Thirty-four patients with long-term (> 3 years) methotrexate use and stress fractures were included in this study. All patient data were evaluated in a retrospective and anonymized design. Data were analyzed according to the rules of the local ethics committee of the University Medical Center Hamburg-Eppendorf, Germany. Skeletal Assessment and Imaging Studies While magnetic resonance imaging (MRI) was performed for individual stress fracture detection, we also determined the areal bone mineral density (aBMD) using dual-energy X-ray absorptiometry (Lunar iDXA; GE Healthcare; Madison, WI, USA) and bone turnover markers from serum and urine samples in all patients. The measured serum markers included 25-hydroxyvitamin D (25-OH-D3), parathyroid hormone (PTH), osteocalcin (Oc), bone-specific alkaline phosphatase (BAP), and deoxypyridinoline cross-links in the urine (DPD). Furthermore, bone microarchitecture was analyzed in 30/34 patients using high-resolution peripheral quantitative computed tomography (HR-pQCT; XtremeCT, Scanco Medical, Switzerland). In the remaining four patients, HR-pQCT could not be performed due to bilateral distal tibia fractures. Scans were performed in the nonfractured distal tibia following a standardized procedure using the standard in vivo patient evaluation protocol [20, 21]. Specifically, we analyzed the trabecular bone mineral density (Tb.BMD), trabecular number (Tb.N), trabecular thickness (Tb.Th) and cortical bone mineral density (Ct.BMD). Stress fractures were also imaged in ten patients using cone beam computed tomography (CBCT) working at 90 kV, 40 mAs with a field of view of 16 × 16 ×  13 cm and slice thickness of 0.2 mm (SCS MedSeries H22, Planmed Oy, Helsinki, Finland). CBCT is a novel imaging technique using divergent X-rays that form a cone, which is increasingly used in extremity imaging, though its initial application was in dentistry and maxillofacial surgery [22, 23]. CBCT enables better fracture detection at extremity sites compared to standard radiography [24]. Furthermore, the spatial resolution of CBCT is higher and the radiation dose exposure is lower compared to conventional multislice CT [25]. Bone Biopsy Studies We obtained four bone biopsies from individuals with long-term MTX treatment and stress fractures. The first biopsy was obtained from an 81-year-old woman suffering from a stress fracture of the proximal right tibia. The biopsy was obtained from the lateral (unaffected) part of the tibial plateau in the course of total knee arthroplasty (TKA) and compared to a biopsy obtained from a 79-year-old female patient with primary osteoarthritis undergoing TKA. The other three biopsies were obtained from fracture sites as a part of a diagnostic work-up in these patients. While the second biopsy was obtained from the distal tibia of a 64-year-old patient previously reported elsewhere [19], the third and fourth biopsies were taken from the distal tibia and the calcaneus of a 71-year-old woman with RA. The specimens were fixed in 3.7% formaldehyde, dehydrated, embedded in methyl-methacrylate, and cut on a Microtec rotation microtome (CVT 4060E, Micro Tec, Walldorf, Germany). Afterwards the 5-µm sections were stained with toluidine blue and von Kossa. Histomorphometric analysis was performed according to the ASBMR nomenclature committee [26]. BV/TV, Tb.N, and Tb.Th as well as osteoid volume per bone volume (OV/BV) were evaluated in the von Kossa-stained sections. The osteoblast surface per bone surface (Ob.S/BS) and the osteoclast surface per bone surface (Oc.S/BS) were evaluated from the toluidine blue-stained sections. Quantitative backscattered electron imaging (qBEI) was performed on the first biopsy using a scanning electron microscope (LEO 435 VP; LEO Electron Microscopy Ltd., Cambridge, England) with a backscattered electron detector (Type 202; K.E. Developments Ltd., Cambridge, England), as described previously [27]. The scanning electron microscope was operated at 20 kV and 680 pA at a constant working distance. The acquired images were analyzed using a customized MATLAB (The MathWorks, Inc., Natick, Massachusetts, USA) script. QBEI was used to measure the bone mineral density distribution based on the generated gray values that represent the mean calcium content (mean Ca-Wt%). For both samples, calcium distribution curves were calculated. Treatment Intervention and Follow-Up Ten patients were evaluated at a follow-up visit at 2.6 ± 1.5 years (min. 1 year, max. 5 years). MTX treatment had been discontinued in 8/10 patients in consultation with a rheumatologist, while the MTX dose was reduced in the remaining 2 patients. Seven out of 10 patients were treated with a subcutaneous administration of denosumab 60 mg (Prolia®, Amgen, USA) every 6 months in combination with a daily subcutaneous administration of teriparatide 20 µg (Forsteo®, Eli Lilly, USA). While two patients were not treated with this bone-specific therapy due to individual contraindications, one patient was treated with teriparatide only. The two patients with no bone-specific treatment had discontinued their MTX medication. All patients received 20,000 IE vitamin D3 and 1 g of dietary calcium daily. At follow-up, clinical reexamination and an HR-pQCT scan were performed. The clinical course was evaluated, and patients were asked about subjective changes in mobility and pain (++/+ major/minor improvement, −/− no improvement/worsened). Furthermore, we performed the Timed Up and Go test and compared the achieved time to previously published reference values [28]. The chair rising test (CRT) was performed as described previously [29], and the results were compared to reference data [30]. Statistical Analysis All data were evaluated using GraphPad Prism® (GraphPad Software, La Jolla, CA, USA). Data are presented as scatter plots with additional labeling of the mean value ± the standard deviation (SD). After checking for normal distribution, the paired t-test was used to compare measurement results from the initial presentation and follow-up. Furthermore, the percent change per year was calculated. P-values of 0.05 or less were considered statistically significant. Results Clinical Characterization A total of 34 patients were evaluated, of whom all had load-dependent pain of the lower limb without adequate trauma in the past. MRI pointed to band- or meander-shaped stress fractures paralleling the former provisional zones of calcification and growth plates in all cases. The most frequent location was the distal tibia (53%) and the calcaneus (53%), followed by the proximal tibia (44%) and the distal femur (18%). In 68% of the patients, we observed stress fractures at multiple and/or bilateral locations (Fig. 1a–e). No association between the time interval of clinical onset and diagnosis and the MTX treatment durations or doses could be detected.Fig. 1 MRI morphology of stress fractures in MTX osteopathy. Band-like stress fractures in proton density (PD)-weighted fat-suppressed (FS) turbo spin echo (TSE) MRI sequences are seen. a Distal tibia, coronal and sagittal plane of two different patients. b Calcaneus, coronal and sagittal plane of two different patients. c Proximal tibia/distal femur, coronal plane. d Multiple stress fractures were found in 68% of the patients. e Bar graph indicating the regional distribution of stress fractures in n = 34 patients (frequency in % for each skeletal site) MTX osteopathy was diagnosed mostly in females (88%, Fig. 2a). The MTX dosage was 18.6 ± 4.9 mg (15–25 mg) weekly. Importantly, a prolonged time between clinical onset and diagnosis of 17.4 ± 8.6 months (5–36 months) was noted. Most patients had a history of corticosteroid treatment; however, upon presentation at our clinic, only 12/34 patients (35.3%) received low-dose oral prednisone (< 5 mg) (Fig. 2b). MTX treatment had been prescribed due to RA (26/34 patients), psoriatic arthritis (4/34 patients), SLE (2/34 patients), polymyalgia rheumatica (1/34 patients) or ankylosing spondylitis (1/34 patients) (Fig. 1c). Only two patients had vertebral fractures. We observed a peak in the occurrence of MTX osteopathy at the age of 70–79 years (Fig. 2d).Fig. 2 Clinical characteristics and bone mineral density, turnover and microstructure. a Eighty-eight percent of the affected patients were female. b Prednisone/no prednisone treatment. c Distribution of the different rheumatic diseases. d Age distribution. e, f DXA T-score at the lumbar spine (LS) and total hip. g, h Serum bone-specific alkaline phosphatase (BAP) and osteocalcin levels (both bone formation) and i urinary deoxypyridinoline (DPD) (bone resorption). Gray boxes indicate reference ranges. j Trabecular number (Tb. N), k trabecular thickness (Tb. Th) and l cortical BMD compared to age- and sex-matched reference data [31] A total of 21/34 (61.8%) of the patients were diagnosed with osteoporosis (i.e., BMD T-score ≤ − 2.5), and the remaining patients were diagnosed with osteopenia, according to the DXA measurements (Fig. 2e, f). Biochemical bone turnover analyses revealed higher levels in the bone-specific alkaline phosphatase than in the osteocalcin levels compared to reference values (Fig. 2g, h). In 18/32 patients (56%), we detected elevated bone resorption markers in the urine (i.e., DPD cross links) (Fig. 2i). HR-pQCT analysis revealed an almost normal trabecular number in most patients and a more severe reduction of trabecular thickness and cortical BMD in the distal tibia compared to reference values (Fig. 2j–l). While two patients presented with 25-hydroxy-vitamin D3 levels < 20 µg/l, ten patients had 25-OH-D3 levels < 30 µg/l. All patients received oral vitamin D supplementation. Cone Beam CT (CBCT) Imaging and Biopsy Findings CBCT imaging demonstrated osseous alterations around the typical stress fracture localizations (distal tibia, around the knee, calcaneus) (Fig. 3a–c). We were able to determine four stages according to the severity of the morphological alterations. Specific findings included epimetaphyseal osteolysis, followed by confluent microcallus formation and band-like sclerosis along the growth plates. These stress fractures were prone to eventually collapsing, leading to severe fractures and deformities in some cases (Supplementary Fig. 1).Fig. 3 Cone beam CT (CBCT) imaging. Differentiation and classification of different disease severities based on CBCT imaging a in the distal tibia, b around the knee (distal femur and proximal tibia) and c in the calcaneus To further characterize the skeletal changes on a microscopic level, a biopsy was obtained from the lateral (nonfractured) part of the tibial plateau. In this patient, the stress fracture of the medial tibial plateau was already visible on conventional radiography (Fig. 4a) and later confirmed by MRI (Fig. 4b). The tibial plateau was subsequently imaged by contact radiography (Fig. 4c). Histological quantification was performed in comparison with an age-matched control (Fig. 4d). The tibial bone microstructure of the patient with MTX osteopathy was characterized by an unchanged BV/TV and a higher trabecular number but a lower trabecular thickness (Fig. 4e–g). Furthermore, a low number of osteoblasts but a high number of osteoclasts were detected, leading to a lower osteoblast surface and a higher osteoclast surface compared to those of the control (Fig. 4h–j). Backscattered electron imaging confirmed highly prevalent eroded surfaces but an overall similar bone mineral density distribution (Fig. 4k, l). Additional histomorphometric analysis of three fracture biopsies from the distal tibiae and the calcaneus revealed the presence of fracture calluses and woven bone but no detection of osteonecrosis. This was associated with a generally high bone turnover (Fig. 5).Fig. 4 Biopsy studies (81-year-old woman) in the lateral (nonfractured) tibial plateau. a Anteroposterior radiograph showing the fracture of the medial tibial plateau (red arrow). b Coronal MRI, PD-weighted sequence. c Contact radiography (lateral view) of the resected tibial plateau, a anterior, p posterior. d Histological overview, von Kossa staining in the patient and the control. e–g Quantification of BV/TV, Tb.N and Tb.Th. h Visible osteoclasts on the surface of a trabecula, toluidine blue staining. i, j Osteoblast and osteoclast surface. k Image obtained by backscattered electron imaging with eroded surfaces (asterisks). l BMDD histograms Fig. 5 Biopsies from different fracture sites reveal interference with fracture healing with chronic callus formation and osteoidosis but an absence of osteonecrosis. a Representative histological images, toluidine blue staining. Left (no. 1): distal tibia (64-year-old woman) showing fracture callus; middle and right (nos. 2, 3): distal tibia and calcaneus (71-year-old woman) with woven bone formation. b Histomorphometric quantification in the individual biopsies nos. 1–3 including osteoid volume per bone volume (OV/BV), osteoblast surface per bone surface (Ob.S/BS) and osteoclast surface per bone surface (Oc.S/BS) Treatment Outcome and Follow-Up MTX treatment was discontinued or replaced in 27/34 patients, while the remaining seven patients underwent MTX dose reduction. The discontinuation of MTX, in consultation with an expert rheumatologist, was well tolerated. MTX was replaced by azathioprine, low-dose glucocorticoids or monoclonal antibodies (e.g., IL-17). Moreover, a bone-specific therapy consisting of denosumab, teriparatide or combined denosumab–teriparatide was initiated on the basis of the risk profiles and pre-existing comorbidities. Follow-up examinations in ten patients (8/10 MTX discontinuation) revealed that no further stress fractures had occurred, while treatment modifications were well tolerated and overall mobility was improved. Most patients showed an adequate increase or at least stable BMD values in the lumbar spine and hip (Table 1). Five of ten patients reported major improvements regarding mobility and pain levels, while the other five patients reported minor improvements (Fig. 6a). Of the 5/10 patients with a major improvement, all had discontinued their MTX medication. The two patients who continued MTX had only minor clinical improvements and showed no increase in BMD. Although MRI follow-up could not be performed in all patients, healing of stress fractures was observed in individual patients who underwent follow-up MRI (Fig. 6b). Successful remobilization and return to daily activities were achieved in all cases. Timed Up and Go testing confirmed the regained mobility, although the reference of 10 s was not reached in most patients (Fig. 6c). The CRT indicated slow but possible rises from a chair, while both tests were not possible in any of the patients at the initial presentation (Fig. 6d). The analysis of bone microarchitecture in the tibia using HR-pQCT revealed constant trabecular parameters and marked improvements in cortical parameters at follow-up (Fig. 6e–j, Supplementary Tab. 1 + 2).Table 1 BMD and corresponding T-scores assessed by DXA in the lumbar spine (LS) and hip of ten patients with follow-up measurements Pat. Time interval (yr.) MTX Specific therapy CC BMD LS T-score LS BMD hip T-score hip Initial Follow-up Change (%) Initial Follow-up Change (SD) Initial Follow -up Change (%) Initial Follow -up Change (SD) 1 1.5 DIS DATA ++ 1.027 1.012 − 1.5 − 1.3 − 1.3 ± 0.0 0.810 0.831 + 2.6 − 1.4 − 1.2 + 0.2 2 2.3 CON DATA + n/a n/a n/a n/a n/a n/a 0.671 0.660 − 1.6 − 3.2 − 3.6 − 0.4 3 4.0 DIS DATA + 0.869 0.957 + 10.1 − 2.8 − 2.0 + 0.8 0.675 0.686 + 1.6 − 2.7 − 2.6 + 0.1 4 4.0 DIS DATA ++ 1.121 1.054 − 6.0 − 0.8 − 0.9 − 0.1 0.689 0.717 + 4.1 − 2.6 − 2.2 + 0.4 5 3.8 DIS DATA ++ 0.709 0.788 + 11.1 − 3.8 − 2.4 + 1.4 0.717 0.726 + 1.3 − 2.4 0.1 + 2.5 6 1.6 DIS no ++ 0.713 0.764 + 7.2 − 4.0 − 3.6 + 0.4 0.609 0.580 − 4.8 − 3.3 − 3.5 − 0.2 7 1.0 DIS DATA ++ 0.950 0.962 + 1.3 − 1.8 − 1.7 + 0.1 0.784 0.797 + 1.7 − 1.6 − 1.5 + 0.1 8 1.6 DIS no + 0.743 0.785 + 5.7 − 3.5 − 3.3 + 0.2 0.660 0.689 + 4.4 − 2.7 − 2.4 + 0.3 9 1.0 DIS DATA + 1.028 1.078 + 4.9 − 1.1 − 0.7 + 0.4 0.779 0.864 + 10.9 − 1.8 − 1.1 + 0.7 10 5.0 CON TPT → D’mab + 0.988 0.961 − 2.7 − 1.8 − 1.8 ± 0.0 n/a n/a n/a n/a n/a n/a Changes are presented as percent (%) or standard deviation (SD) Yr. years, DIS discontinued, CON continued, DATA denosumab and teriparatide, D’mab denosumab, TPT teriparatide, CC clinical course (++/+ major/minor clinical improvement), n/a not available due to previous orthopedic surgery such as spondylodesis or hip replacement Fig. 6 Follow-up and treatment response. a Subjective clinical course (++ major improvement, + minor improvement, − no improvement in mobility and pain). b MRI, T1-weighted, sagittal sequence. c Individual results for the Timed Up and Go test at follow-up. d Chair rising test (CRT). e–j HR-pQCT at initial presentation and follow-up (distal tibia for all panels). e Trabecular BMD. f Trabecular number. g Cortical BMD. h Cortical thickness. i HR-pQCT image at 0 and 14 months indicating increasing cortical thickness (white arrows). j HR-pQCT changes converted to %-change/1 year Discussion While stress fractures in patients with long-term MTX use have been repeatedly described, detailed knowledge on the underlying skeletal alterations in a larger patient collective was not available to date. From a clinical perspective, our results suggest that long-term MTX treatment may have negative effects on bone metabolism and quality in certain patient groups, which was expressed by stress fractures with a unique band- or meander-shaped appearance along the growth plate. Indeed, our MRI-based morphological characterization therefore corresponds well to the first reports on MTX osteopathy [3, 4, 6]. In other words, this comprehensive imaging analysis illustrates that the detected uniquely shaped stress fractures constitute the hallmark of MTX osteopathy. Even in different rheumatic diseases, this typical clinical and radiological picture was detected, emphasizing the concept of significant MTX side effects rather than concomitant phenomena of the underlying diseases. At the same time, stress fractures due to other reasons including overuse, osteomalacia (e.g., calcium malabsorption, phosphate wasting) or inflammatory conditions are typically not characterized by a meander-shaped morphology along the growth plates. The skeletal alterations were subsequently characterized by CBCT, where a consistent radiographic pattern of epimetaphyseal osteolysis and band-like sclerosis (i.e., microcallus) was found. In association with this pathognomonic imaging appearance detected by MRI and CBCT, we observed a consistent bone microstructure and turnover pattern. Namely, the skeletal deterioration was further characterized by HR-pQCT, where pronounced trabecular thinning was detected. Bone turnover was often characterized by a combination of high bone-specific alkaline phosphatase levels, low osteocalcin levels indicating low bone formation, and elevated bone resorption parameters (i.e., urinary DPD cross links). This dissociation between low bone formation and high bone resorption was recapitulated in a bone biopsy obtained from a patient with MTX osteopathy, while three biopsies from fracture sites indicated interference with fracture healing but an absence of osteonecrosis. In sum, the detected bone turnover dissociation is most likely a major contributing factor for the detected long-term skeletal complications. Based on our findings, we suggest that any symptomatic patient with lower extremity pain, loss of mobility and long-term MTX treatment should be rigorously screened for stress fractures and skeletal status (i.e., osteoporosis) using MRI, DXA and laboratory analyses. CBCT represents a valuable tool to further image these osseous alterations at a high resolution while simultaneously reducing radiation exposure [22]. While the correct diagnosis can help to ensure the best medical care in terms of appropriate treatment, there is a risk that stress fractures will progress towards extensive bone and joint destruction (i.e., collapse) if not treated properly and MTX is continued in these patients (Supplementary Fig. 1). Although we do understand that a discontinuation of MTX might not be feasible in all patients with different rheumatic inflammatory diseases, MTX discontinuation should always be considered in patients with MTX osteopathy. As the diagnosed stress fractures were accompanied by osteoporosis or osteopenia, according to DXA, and a dissociation of low bone formation and elevated bone resorption in most cases, we treated most our patients with both denosumab (anti-resorptive) and teriparatide (osteoanabolic) according to the previously published DATA study [32]. We additionally guaranteed sufficient 25-OH-D3 levels, as this was found to be beneficial for reducing the risk of stress fractures [33]. In combination with a discontinuation of MTX treatment, the analyzed subset of ten patients recovered from stress fractures and regained their mobility, which was associated with increased BMD levels and improved cortical microstructure in most patients. Close rheumatologic monitoring was carried out to avoid flares of the underlying autoimmune disease. The denosumab–teriparatide combination therapy was previously shown to lead to the highest increases in BMD [32] before the sclerostin antibody romosozumab entered clinical research [34]. However, it is currently not known whether these findings can be applied to special patient groups such as RA patients with MTX osteopathy. In rats with MTX-induced osteopathy, anti-resorptive treatment was found to be effective [11]. The addition of denosumab to MTX has also been found to be an effective treatment option in the prevention of further joint destruction in patients with RA [35]. We are aware that our study only offers correlative evidence regarding MTX osteopathy as a clinical entity that is described by pathognomonic and nonself-limiting stress fractures. Our results do not provide sufficient information in terms of causality and stress fracture prevalence due to long-term MTX treatment, and future studies will have to elaborate on the epidemiologic aspects of this burden. Our data also do not allow us to determine whether MTX discontinuation or the initiated bone-specific therapy is more effective regarding the achieved clinical improvements. It is interesting to note that from the ten patients analyzed at follow-up, the two patients who continued MTX showed minor clinical improvements and no increase in BMD. Finally, other conditions, such as inflammation, corticosteroid use or postmenopausal status, also promote an impairment in bone health. Nonetheless, several studies have linked MTX to deteriorated bone quality. Reduced BMD was observed with MTX use compared to no MTX use among patients with RA [36]. In a prospective trial of 329 RA patients, a negative annual BMD change was significantly associated with MTX dosage [37]. Moreover, even in several other reported cases of patients with RA and stress fractures, most of them had previously undergone long-term MTX therapy [38–40]. On the other hand, MTX was associated with neither improved nor worsened bone microstructure in patients with PsA, while DMARD use (such as TNFalpha inhibitors or IL-17A antibodies) led to improved microstructure [41]. In general, it remains a major issue to develop optimal drug strategies for treating bone problems in patients with inflammatory diseases [42]. Taken together, we have provided clinical evidence of the potential negative effects of long-term MTX use on bone quality, associated with peculiar stress fractures that can be positively influenced by a discontinuation of MTX together with the initiation of a combined denosumab and teriparatide treatment. Whether the observed improvements in stress fractures and changes in quality translate into reductions in stress fracture risk over the long term remains to be evaluated in larger series. Electronic supplementary material Below is the link to the electronic supplementary material.Supplementary file1 (PDF 1308 kb) Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Tim Rolvien and Nico Maximilian Jandl contributed equally to this work. Acknowledgements The authors thank Andrea Thieke, Olga Winter and Elke Leicht for technical assistance with the histological sections. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with Ethical Standards Conflict of interest Tim Rolvien, Nico Maximilian Jandl, Julian Stürznickel, Frank Timo Beil, Ina Kötter, Ralf Oheim, Ansgar W Lohse, Florian Barvencik and Michael Amling declare that they have no conflict of interest. Human and Animal Rights All procedures are in accordance with the ethical standards as laid down in the 1964 Declaration of Helsinki and its later amendments or comparable ethical standards. Informed Consent Informed consent was obtained from all individual patients included in this study.
Recovering
ReactionOutcome
CC BY
33064170
18,451,574
2021-02
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Torsade de pointes'.
Torsade de pointes induced by intravenous amiodarone therapy accompanied by marked augmentation of the transmural dispersion of repolarization in a patient with tachycardia-induced-cardiomyopathy. We report a 77-year-old human on renal dialysis for end-stage renal disease with heart failure and atrial fibrillation (AF) complicated by a high ventricular frequency. The underlying disease was thought as tachycardia-induced-cardiomyopathy. Intravenous infusion of amiodarone was initiated, and direct current cardioversion succeeded in converting AF to sinus rhythm. Then, excessive increases in the QT and Tpeak-Tend (Tp-e) intervals were seen and hypokalemia induced by hemodialysis led to the development of numerous episodes of torsades de pointes (TdP). Magnesium repletion was effective in preventing TdP, while Tp-e intervals returned to the previous values 2 days after the discontinuation of amiodarone. 1 CASE REPORT A 77‐year‐old human on regular hemodialysis for end‐stage renal disease was admitted because of heart failure. He had been well until persistent atrial fibrillation (AF) with rapid ventricular response started 2 months prior. His past medical history included gastric cancer and bile duct cancer surgeries. ECG on admission revealed AF with a heart rate in the 100 s, and poor R wave progression with newly developed negative T waves in the precordial leads (Figure 1b), however, coronary angiography revealed no stenosis. Chest radiography confirmed left‐sided pleural effusion. Transthoracic echocardiography revealed diffuse left ventricular (LV) hypokinesis, with ejection fraction of 25%, which was 58% 3 months prior during sinus rhythm (SR). As the rapid AF was sustained and there were no other causes of the LV dysfunction, tachycardia‐induced‐LV dysfunction and heart failure were suspected. Transesophageal echocardiography revealed no intracardiac thrombus; then, intravenous amiodarone was initiated, and DC cardioversion succeeded in converting the AF to SR (Figure 1c). The QT intervals were measured manually with calipers in all 12 leads. They were defined as the time interval between the earliest deflection of the QRS complex and the point of T‐wave offset, which was defined by the return of the terminal T wave to the isoelectric baseline. When U waves were present, U waves were excluded using the presented guidelines (Lepechkin & Surawicz, 1952), therefore, QT interval was measured to the nadir of the curve between the T wave and U wave. Biphasic T waves were distinguished from U waves by comparison with similar complexes in contiguous ECG leads. If the end of the T wave could not be reliably determined or when the T waves were isoelectric or of low amplitude, QT measurements were not made and these leads were excluded from analysis. Then, T‐wave peak was determined, and the Tpeak‐Tend (Tp‐e) intervals from T‐wave peak to T‐wave end were measured. In the case of negative or biphasic T waves, T‐wave peak was defined to the nadir of the T wave. T waves smaller than 1.5 mm in amplitude were not measured. The Tp‐e value reported was the maximum. All measured QT and Tp‐e intervals were corrected (QTc and c‐Tp‐e) for heart rate using the Bazett formula (QTc; QT/√RR, c‐Tp‐e; Tp‐e/√RR). QTc interval was 444 ms during AF on admission, and after the conversion to SR, QTc interval was 440 ms and c‐Tp‐e 42 ms. FIGURE 1 The serial ECGs from 6 months prior to admission (a), on admission (b), just after successful direct current cardioversion with 4 hr of administration of intravenous amiodarone (c), and 15 hr after beginning amiodarone (d). (a) The rhythm was sinus rhythm (SR) with QTc interval of 426 ms. T waves were positive and symmetrical, and poor R wave progression was seen. (b) The rhythm was atrial fibrillation with a heart rate in the 100 s and QTc interval of 444 ms. Negative T waves in precordial leads newly developed. (c) Just after the recovery to SR, QTc interval was 440 ms. Negative T waves in the precordial leads were symmetrical with Tpeak‐Tend (Tp‐e) interval of 40 ms and corrected‐Tp‐e (c‐Tp‐e) interval of 42 ms. (d) QTc interval prolonged to 517 ms, which was accompanied by prolonged Tp‐e interval of 240 ms (c‐Tp‐e interval: 255 ms) ECG taken 15 hr after beginning amiodarone revealed that QTc interval prolonged of 517 ms, which was accompanied by prolonged c‐Tp‐e interval of 255 ms (Figure 1d), with normal serum electrolytes. Therefore, the intravenous amiodarone was ceased (total dose of 370 mg). However, QTc and c‐Tp‐e interval did not recover; on the contrary, both prolonged even further after hemodialysis, which triggered hypokalemia (3.3 mEq/L). The patient developed repetitive short‐lasting torsade de pointes tachycardias (TdPs) terminating spontaneously (Figure 2). During the consecutive TdP episodes, he had syncope once. Magnesium repletion was effective in preventing TdP; however, T waves became symmetrical and c‐Tp‐e intervals shortened 2 days after the discontinuation of amiodarone (Figure 3). During the TdP episodes, isoproterenol could not be administered for fear of inducing AF. Following catheter ablation of AF, he kept SR. No TdP recurred, and the patient remained asymptomatic. Three months later, catheter ablation of atrial tachycardia which occurred newly was performed. LV function recovered 8 months later. FIGURE 2 12‐lead ECG (a) and ECG monitor strips (b) after hemodialysis. After regular hemodialysis, mild hypokalemia was induced. (a) Note the remarkably bizarre negative T waves in the precordial leads, which did not end when the next sinus rhythm QRS complex started. Therefore, QT or Tp‐e intervals could not be measured. In the 5th beat, torsade de pointes (TdP) terminated spontaneously. (b) The patient developed repetitive short‐lasting TdP episodes terminating spontaneously. A “short‐long‐short” sequence preceded the onset of TdP FIGURE 3 The serial ECGs from 10 hr after the discontinuation of amiodarone (a), 1 day after the discontinuation (b), 2 days after the discontinuation (c), and 1 month later (d). (a, b) T waves in the precordial leads were still negative and corrected‐ Tpeak‐Tend (c‐Tp‐e interval) was prolonged (291 and 341 ms, each). (c) Negative T waves were seen in all chest leads; however, they were symmetrical. (d) T waves in all chest leads were positive and symmetrical 2 DISCUSSION Amiodarone is widely used for the treatment of malignant arrhythmias with a remarkably low frequency of proarrhythmia. The incidence of TdP associated with oral amiodarone is reported to be <1.0% (Hohnloser et al., 1994), while that with intravenous amiodarone is about 1.5% (Shenthar et al., 2017). Although both amiodarone and other antiarrhythmic drugs including class Ia and other class III prolong the QT interval, TdP is overwhelmingly rare during amiodarone therapy. The mechanism of the difference is that amiodarone homogeneously prolongs the ventricular repolarization, whereas other antiarrhythmic drugs prolong it in a nonhomogeneous fashion accompanied by an increase in QT interval dispersion (Friedman & Stevenson, 1998; Hii et al., 1992; Milberg et al., 2004; van Opstal et al., 2001). Antezelvich introduced the concept of Tp‐e interval in surface ECG as an index of transmural dispersion of repolarization (TDR) based on the studies with the coronary‐perfused wedge preparation, that repolarization of the epicardial action potential coincides with the peak of the T wave and repolarization of the mid‐myocardial cells (M cells) is coincident with the end of the T wave, so that Tp‐e interval provides a measure of TDR, with forecasting risk for the development of TdP (Sicouri & Antzelevitch, 1991). Studies have indicated that drugs that do not increase TDR have little or no potential to induce TdP despite causing a prolongation of the QT interval. Amiodarone has in common the ability to block IKs, IKr, and late INa. This combination produces a preferential prolongation of APD of the epicardium and endocardium so that the QT interval is prolonged, but the TDR is actually reduced and TdP rarely, if ever, occurs under these conditions (Hii et al., 1992; Kotake et al., 2015; Milberg et al., 2004; van Opstal et al., 2001). The case reports of amiodarone‐induced‐TdP so far were rare and have shown significant QT prolongation, but without Tp‐e prolongation (Belardinelli et al., 2003; Friedman & Stevenson, 1998; Hii et al., 1992; Hohnloser et al., 1994; Kotake et al., 2015; Shenthar et al., 2017). The increased TDR could provide a more accurate electrophysiologic marker of the risk for TdP than does the QT interval. In this case with tachycardia‐induced‐cardiomyopathy (TCM) due to rapid AF, the QT interval prolonged dramatically with bizarre T waves suggesting augmented TDR after the administration of intravenous amiodarone. Finally, hypokalemia caused by hemodialysis initiated TdP accompanied by syncope. To the best of our knowledge, this is the first report of TdP with marked Tp‐e prolongation in a patient during amiodarone therapy. Although the mechanism responsible for the marked augmentation of TDR with amiodarone in our case is unclear, it would be speculated as multifactorial. TDR is increased by drugs, heart failure, acute myocardial infarction, and various channelopathies. First, we previously reported the possibility that the transmural heterogeneity of myocardial ischemia might influence the repolarization resulting in increased TDR (Kawabata et al., 2008). Although CAG revealed no lesions in this case, the possibility of myocardial ischemia in microvascular level could not be completely ruled out. Second, as both QT and Tp‐e intervals were not prolonged before the administration of amiodarone in this case, the marked QT/TDR increase was induced by amiodarone; however, the possibility of acquired long QT syndrome based on the genetic causes could not be excluded as a genetic test was not performed. Third, the underlying disease was thought as TCM in this patient. It is reported that in tachycardia‐induced heart failure K+ currents are down‐regulated ununiformly, causing enhanced TDR (Akar et al., 2005). It was speculated that the underlying molecular features in TCM would influence the augmentation of TDR in this case. Forth, a different distribution or clearance of amiodarone could be related. In previous report of animal models of acquired long QT syndrome, amiodarone increased QTc time in 6 of 7 dogs, while dispersion of repolarization was increased in 3. The three dogs tended to have higher tissue levels of amiodarone and its metabolite compared with those without dispersion of repolarization (van Opstal et al., 2001). In this case, we did not check the concentration of amiodarone or n‐desethylamiodarone; however, there was a possibility that they were quite high. 3 CONCLUSION Although the reported incidence of TdP during amiodarone therapy is low, careful ECG monitoring should be undergone to check not only QT interval but Tp‐e interval. CONFLICT OF INTEREST None. ETHICAL APPROVAL The authors have obtained the patient's informed consent. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
AMIODARONE HYDROCHLORIDE
DrugsGivenReaction
CC BY
33070441
18,469,623
2021-05
What was the administration route of drug 'AMIODARONE HYDROCHLORIDE'?
Torsade de pointes induced by intravenous amiodarone therapy accompanied by marked augmentation of the transmural dispersion of repolarization in a patient with tachycardia-induced-cardiomyopathy. We report a 77-year-old human on renal dialysis for end-stage renal disease with heart failure and atrial fibrillation (AF) complicated by a high ventricular frequency. The underlying disease was thought as tachycardia-induced-cardiomyopathy. Intravenous infusion of amiodarone was initiated, and direct current cardioversion succeeded in converting AF to sinus rhythm. Then, excessive increases in the QT and Tpeak-Tend (Tp-e) intervals were seen and hypokalemia induced by hemodialysis led to the development of numerous episodes of torsades de pointes (TdP). Magnesium repletion was effective in preventing TdP, while Tp-e intervals returned to the previous values 2 days after the discontinuation of amiodarone. 1 CASE REPORT A 77‐year‐old human on regular hemodialysis for end‐stage renal disease was admitted because of heart failure. He had been well until persistent atrial fibrillation (AF) with rapid ventricular response started 2 months prior. His past medical history included gastric cancer and bile duct cancer surgeries. ECG on admission revealed AF with a heart rate in the 100 s, and poor R wave progression with newly developed negative T waves in the precordial leads (Figure 1b), however, coronary angiography revealed no stenosis. Chest radiography confirmed left‐sided pleural effusion. Transthoracic echocardiography revealed diffuse left ventricular (LV) hypokinesis, with ejection fraction of 25%, which was 58% 3 months prior during sinus rhythm (SR). As the rapid AF was sustained and there were no other causes of the LV dysfunction, tachycardia‐induced‐LV dysfunction and heart failure were suspected. Transesophageal echocardiography revealed no intracardiac thrombus; then, intravenous amiodarone was initiated, and DC cardioversion succeeded in converting the AF to SR (Figure 1c). The QT intervals were measured manually with calipers in all 12 leads. They were defined as the time interval between the earliest deflection of the QRS complex and the point of T‐wave offset, which was defined by the return of the terminal T wave to the isoelectric baseline. When U waves were present, U waves were excluded using the presented guidelines (Lepechkin & Surawicz, 1952), therefore, QT interval was measured to the nadir of the curve between the T wave and U wave. Biphasic T waves were distinguished from U waves by comparison with similar complexes in contiguous ECG leads. If the end of the T wave could not be reliably determined or when the T waves were isoelectric or of low amplitude, QT measurements were not made and these leads were excluded from analysis. Then, T‐wave peak was determined, and the Tpeak‐Tend (Tp‐e) intervals from T‐wave peak to T‐wave end were measured. In the case of negative or biphasic T waves, T‐wave peak was defined to the nadir of the T wave. T waves smaller than 1.5 mm in amplitude were not measured. The Tp‐e value reported was the maximum. All measured QT and Tp‐e intervals were corrected (QTc and c‐Tp‐e) for heart rate using the Bazett formula (QTc; QT/√RR, c‐Tp‐e; Tp‐e/√RR). QTc interval was 444 ms during AF on admission, and after the conversion to SR, QTc interval was 440 ms and c‐Tp‐e 42 ms. FIGURE 1 The serial ECGs from 6 months prior to admission (a), on admission (b), just after successful direct current cardioversion with 4 hr of administration of intravenous amiodarone (c), and 15 hr after beginning amiodarone (d). (a) The rhythm was sinus rhythm (SR) with QTc interval of 426 ms. T waves were positive and symmetrical, and poor R wave progression was seen. (b) The rhythm was atrial fibrillation with a heart rate in the 100 s and QTc interval of 444 ms. Negative T waves in precordial leads newly developed. (c) Just after the recovery to SR, QTc interval was 440 ms. Negative T waves in the precordial leads were symmetrical with Tpeak‐Tend (Tp‐e) interval of 40 ms and corrected‐Tp‐e (c‐Tp‐e) interval of 42 ms. (d) QTc interval prolonged to 517 ms, which was accompanied by prolonged Tp‐e interval of 240 ms (c‐Tp‐e interval: 255 ms) ECG taken 15 hr after beginning amiodarone revealed that QTc interval prolonged of 517 ms, which was accompanied by prolonged c‐Tp‐e interval of 255 ms (Figure 1d), with normal serum electrolytes. Therefore, the intravenous amiodarone was ceased (total dose of 370 mg). However, QTc and c‐Tp‐e interval did not recover; on the contrary, both prolonged even further after hemodialysis, which triggered hypokalemia (3.3 mEq/L). The patient developed repetitive short‐lasting torsade de pointes tachycardias (TdPs) terminating spontaneously (Figure 2). During the consecutive TdP episodes, he had syncope once. Magnesium repletion was effective in preventing TdP; however, T waves became symmetrical and c‐Tp‐e intervals shortened 2 days after the discontinuation of amiodarone (Figure 3). During the TdP episodes, isoproterenol could not be administered for fear of inducing AF. Following catheter ablation of AF, he kept SR. No TdP recurred, and the patient remained asymptomatic. Three months later, catheter ablation of atrial tachycardia which occurred newly was performed. LV function recovered 8 months later. FIGURE 2 12‐lead ECG (a) and ECG monitor strips (b) after hemodialysis. After regular hemodialysis, mild hypokalemia was induced. (a) Note the remarkably bizarre negative T waves in the precordial leads, which did not end when the next sinus rhythm QRS complex started. Therefore, QT or Tp‐e intervals could not be measured. In the 5th beat, torsade de pointes (TdP) terminated spontaneously. (b) The patient developed repetitive short‐lasting TdP episodes terminating spontaneously. A “short‐long‐short” sequence preceded the onset of TdP FIGURE 3 The serial ECGs from 10 hr after the discontinuation of amiodarone (a), 1 day after the discontinuation (b), 2 days after the discontinuation (c), and 1 month later (d). (a, b) T waves in the precordial leads were still negative and corrected‐ Tpeak‐Tend (c‐Tp‐e interval) was prolonged (291 and 341 ms, each). (c) Negative T waves were seen in all chest leads; however, they were symmetrical. (d) T waves in all chest leads were positive and symmetrical 2 DISCUSSION Amiodarone is widely used for the treatment of malignant arrhythmias with a remarkably low frequency of proarrhythmia. The incidence of TdP associated with oral amiodarone is reported to be <1.0% (Hohnloser et al., 1994), while that with intravenous amiodarone is about 1.5% (Shenthar et al., 2017). Although both amiodarone and other antiarrhythmic drugs including class Ia and other class III prolong the QT interval, TdP is overwhelmingly rare during amiodarone therapy. The mechanism of the difference is that amiodarone homogeneously prolongs the ventricular repolarization, whereas other antiarrhythmic drugs prolong it in a nonhomogeneous fashion accompanied by an increase in QT interval dispersion (Friedman & Stevenson, 1998; Hii et al., 1992; Milberg et al., 2004; van Opstal et al., 2001). Antezelvich introduced the concept of Tp‐e interval in surface ECG as an index of transmural dispersion of repolarization (TDR) based on the studies with the coronary‐perfused wedge preparation, that repolarization of the epicardial action potential coincides with the peak of the T wave and repolarization of the mid‐myocardial cells (M cells) is coincident with the end of the T wave, so that Tp‐e interval provides a measure of TDR, with forecasting risk for the development of TdP (Sicouri & Antzelevitch, 1991). Studies have indicated that drugs that do not increase TDR have little or no potential to induce TdP despite causing a prolongation of the QT interval. Amiodarone has in common the ability to block IKs, IKr, and late INa. This combination produces a preferential prolongation of APD of the epicardium and endocardium so that the QT interval is prolonged, but the TDR is actually reduced and TdP rarely, if ever, occurs under these conditions (Hii et al., 1992; Kotake et al., 2015; Milberg et al., 2004; van Opstal et al., 2001). The case reports of amiodarone‐induced‐TdP so far were rare and have shown significant QT prolongation, but without Tp‐e prolongation (Belardinelli et al., 2003; Friedman & Stevenson, 1998; Hii et al., 1992; Hohnloser et al., 1994; Kotake et al., 2015; Shenthar et al., 2017). The increased TDR could provide a more accurate electrophysiologic marker of the risk for TdP than does the QT interval. In this case with tachycardia‐induced‐cardiomyopathy (TCM) due to rapid AF, the QT interval prolonged dramatically with bizarre T waves suggesting augmented TDR after the administration of intravenous amiodarone. Finally, hypokalemia caused by hemodialysis initiated TdP accompanied by syncope. To the best of our knowledge, this is the first report of TdP with marked Tp‐e prolongation in a patient during amiodarone therapy. Although the mechanism responsible for the marked augmentation of TDR with amiodarone in our case is unclear, it would be speculated as multifactorial. TDR is increased by drugs, heart failure, acute myocardial infarction, and various channelopathies. First, we previously reported the possibility that the transmural heterogeneity of myocardial ischemia might influence the repolarization resulting in increased TDR (Kawabata et al., 2008). Although CAG revealed no lesions in this case, the possibility of myocardial ischemia in microvascular level could not be completely ruled out. Second, as both QT and Tp‐e intervals were not prolonged before the administration of amiodarone in this case, the marked QT/TDR increase was induced by amiodarone; however, the possibility of acquired long QT syndrome based on the genetic causes could not be excluded as a genetic test was not performed. Third, the underlying disease was thought as TCM in this patient. It is reported that in tachycardia‐induced heart failure K+ currents are down‐regulated ununiformly, causing enhanced TDR (Akar et al., 2005). It was speculated that the underlying molecular features in TCM would influence the augmentation of TDR in this case. Forth, a different distribution or clearance of amiodarone could be related. In previous report of animal models of acquired long QT syndrome, amiodarone increased QTc time in 6 of 7 dogs, while dispersion of repolarization was increased in 3. The three dogs tended to have higher tissue levels of amiodarone and its metabolite compared with those without dispersion of repolarization (van Opstal et al., 2001). In this case, we did not check the concentration of amiodarone or n‐desethylamiodarone; however, there was a possibility that they were quite high. 3 CONCLUSION Although the reported incidence of TdP during amiodarone therapy is low, careful ECG monitoring should be undergone to check not only QT interval but Tp‐e interval. CONFLICT OF INTEREST None. ETHICAL APPROVAL The authors have obtained the patient's informed consent. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33070441
18,469,623
2021-05
What was the outcome of reaction 'Torsade de pointes'?
Torsade de pointes induced by intravenous amiodarone therapy accompanied by marked augmentation of the transmural dispersion of repolarization in a patient with tachycardia-induced-cardiomyopathy. We report a 77-year-old human on renal dialysis for end-stage renal disease with heart failure and atrial fibrillation (AF) complicated by a high ventricular frequency. The underlying disease was thought as tachycardia-induced-cardiomyopathy. Intravenous infusion of amiodarone was initiated, and direct current cardioversion succeeded in converting AF to sinus rhythm. Then, excessive increases in the QT and Tpeak-Tend (Tp-e) intervals were seen and hypokalemia induced by hemodialysis led to the development of numerous episodes of torsades de pointes (TdP). Magnesium repletion was effective in preventing TdP, while Tp-e intervals returned to the previous values 2 days after the discontinuation of amiodarone. 1 CASE REPORT A 77‐year‐old human on regular hemodialysis for end‐stage renal disease was admitted because of heart failure. He had been well until persistent atrial fibrillation (AF) with rapid ventricular response started 2 months prior. His past medical history included gastric cancer and bile duct cancer surgeries. ECG on admission revealed AF with a heart rate in the 100 s, and poor R wave progression with newly developed negative T waves in the precordial leads (Figure 1b), however, coronary angiography revealed no stenosis. Chest radiography confirmed left‐sided pleural effusion. Transthoracic echocardiography revealed diffuse left ventricular (LV) hypokinesis, with ejection fraction of 25%, which was 58% 3 months prior during sinus rhythm (SR). As the rapid AF was sustained and there were no other causes of the LV dysfunction, tachycardia‐induced‐LV dysfunction and heart failure were suspected. Transesophageal echocardiography revealed no intracardiac thrombus; then, intravenous amiodarone was initiated, and DC cardioversion succeeded in converting the AF to SR (Figure 1c). The QT intervals were measured manually with calipers in all 12 leads. They were defined as the time interval between the earliest deflection of the QRS complex and the point of T‐wave offset, which was defined by the return of the terminal T wave to the isoelectric baseline. When U waves were present, U waves were excluded using the presented guidelines (Lepechkin & Surawicz, 1952), therefore, QT interval was measured to the nadir of the curve between the T wave and U wave. Biphasic T waves were distinguished from U waves by comparison with similar complexes in contiguous ECG leads. If the end of the T wave could not be reliably determined or when the T waves were isoelectric or of low amplitude, QT measurements were not made and these leads were excluded from analysis. Then, T‐wave peak was determined, and the Tpeak‐Tend (Tp‐e) intervals from T‐wave peak to T‐wave end were measured. In the case of negative or biphasic T waves, T‐wave peak was defined to the nadir of the T wave. T waves smaller than 1.5 mm in amplitude were not measured. The Tp‐e value reported was the maximum. All measured QT and Tp‐e intervals were corrected (QTc and c‐Tp‐e) for heart rate using the Bazett formula (QTc; QT/√RR, c‐Tp‐e; Tp‐e/√RR). QTc interval was 444 ms during AF on admission, and after the conversion to SR, QTc interval was 440 ms and c‐Tp‐e 42 ms. FIGURE 1 The serial ECGs from 6 months prior to admission (a), on admission (b), just after successful direct current cardioversion with 4 hr of administration of intravenous amiodarone (c), and 15 hr after beginning amiodarone (d). (a) The rhythm was sinus rhythm (SR) with QTc interval of 426 ms. T waves were positive and symmetrical, and poor R wave progression was seen. (b) The rhythm was atrial fibrillation with a heart rate in the 100 s and QTc interval of 444 ms. Negative T waves in precordial leads newly developed. (c) Just after the recovery to SR, QTc interval was 440 ms. Negative T waves in the precordial leads were symmetrical with Tpeak‐Tend (Tp‐e) interval of 40 ms and corrected‐Tp‐e (c‐Tp‐e) interval of 42 ms. (d) QTc interval prolonged to 517 ms, which was accompanied by prolonged Tp‐e interval of 240 ms (c‐Tp‐e interval: 255 ms) ECG taken 15 hr after beginning amiodarone revealed that QTc interval prolonged of 517 ms, which was accompanied by prolonged c‐Tp‐e interval of 255 ms (Figure 1d), with normal serum electrolytes. Therefore, the intravenous amiodarone was ceased (total dose of 370 mg). However, QTc and c‐Tp‐e interval did not recover; on the contrary, both prolonged even further after hemodialysis, which triggered hypokalemia (3.3 mEq/L). The patient developed repetitive short‐lasting torsade de pointes tachycardias (TdPs) terminating spontaneously (Figure 2). During the consecutive TdP episodes, he had syncope once. Magnesium repletion was effective in preventing TdP; however, T waves became symmetrical and c‐Tp‐e intervals shortened 2 days after the discontinuation of amiodarone (Figure 3). During the TdP episodes, isoproterenol could not be administered for fear of inducing AF. Following catheter ablation of AF, he kept SR. No TdP recurred, and the patient remained asymptomatic. Three months later, catheter ablation of atrial tachycardia which occurred newly was performed. LV function recovered 8 months later. FIGURE 2 12‐lead ECG (a) and ECG monitor strips (b) after hemodialysis. After regular hemodialysis, mild hypokalemia was induced. (a) Note the remarkably bizarre negative T waves in the precordial leads, which did not end when the next sinus rhythm QRS complex started. Therefore, QT or Tp‐e intervals could not be measured. In the 5th beat, torsade de pointes (TdP) terminated spontaneously. (b) The patient developed repetitive short‐lasting TdP episodes terminating spontaneously. A “short‐long‐short” sequence preceded the onset of TdP FIGURE 3 The serial ECGs from 10 hr after the discontinuation of amiodarone (a), 1 day after the discontinuation (b), 2 days after the discontinuation (c), and 1 month later (d). (a, b) T waves in the precordial leads were still negative and corrected‐ Tpeak‐Tend (c‐Tp‐e interval) was prolonged (291 and 341 ms, each). (c) Negative T waves were seen in all chest leads; however, they were symmetrical. (d) T waves in all chest leads were positive and symmetrical 2 DISCUSSION Amiodarone is widely used for the treatment of malignant arrhythmias with a remarkably low frequency of proarrhythmia. The incidence of TdP associated with oral amiodarone is reported to be <1.0% (Hohnloser et al., 1994), while that with intravenous amiodarone is about 1.5% (Shenthar et al., 2017). Although both amiodarone and other antiarrhythmic drugs including class Ia and other class III prolong the QT interval, TdP is overwhelmingly rare during amiodarone therapy. The mechanism of the difference is that amiodarone homogeneously prolongs the ventricular repolarization, whereas other antiarrhythmic drugs prolong it in a nonhomogeneous fashion accompanied by an increase in QT interval dispersion (Friedman & Stevenson, 1998; Hii et al., 1992; Milberg et al., 2004; van Opstal et al., 2001). Antezelvich introduced the concept of Tp‐e interval in surface ECG as an index of transmural dispersion of repolarization (TDR) based on the studies with the coronary‐perfused wedge preparation, that repolarization of the epicardial action potential coincides with the peak of the T wave and repolarization of the mid‐myocardial cells (M cells) is coincident with the end of the T wave, so that Tp‐e interval provides a measure of TDR, with forecasting risk for the development of TdP (Sicouri & Antzelevitch, 1991). Studies have indicated that drugs that do not increase TDR have little or no potential to induce TdP despite causing a prolongation of the QT interval. Amiodarone has in common the ability to block IKs, IKr, and late INa. This combination produces a preferential prolongation of APD of the epicardium and endocardium so that the QT interval is prolonged, but the TDR is actually reduced and TdP rarely, if ever, occurs under these conditions (Hii et al., 1992; Kotake et al., 2015; Milberg et al., 2004; van Opstal et al., 2001). The case reports of amiodarone‐induced‐TdP so far were rare and have shown significant QT prolongation, but without Tp‐e prolongation (Belardinelli et al., 2003; Friedman & Stevenson, 1998; Hii et al., 1992; Hohnloser et al., 1994; Kotake et al., 2015; Shenthar et al., 2017). The increased TDR could provide a more accurate electrophysiologic marker of the risk for TdP than does the QT interval. In this case with tachycardia‐induced‐cardiomyopathy (TCM) due to rapid AF, the QT interval prolonged dramatically with bizarre T waves suggesting augmented TDR after the administration of intravenous amiodarone. Finally, hypokalemia caused by hemodialysis initiated TdP accompanied by syncope. To the best of our knowledge, this is the first report of TdP with marked Tp‐e prolongation in a patient during amiodarone therapy. Although the mechanism responsible for the marked augmentation of TDR with amiodarone in our case is unclear, it would be speculated as multifactorial. TDR is increased by drugs, heart failure, acute myocardial infarction, and various channelopathies. First, we previously reported the possibility that the transmural heterogeneity of myocardial ischemia might influence the repolarization resulting in increased TDR (Kawabata et al., 2008). Although CAG revealed no lesions in this case, the possibility of myocardial ischemia in microvascular level could not be completely ruled out. Second, as both QT and Tp‐e intervals were not prolonged before the administration of amiodarone in this case, the marked QT/TDR increase was induced by amiodarone; however, the possibility of acquired long QT syndrome based on the genetic causes could not be excluded as a genetic test was not performed. Third, the underlying disease was thought as TCM in this patient. It is reported that in tachycardia‐induced heart failure K+ currents are down‐regulated ununiformly, causing enhanced TDR (Akar et al., 2005). It was speculated that the underlying molecular features in TCM would influence the augmentation of TDR in this case. Forth, a different distribution or clearance of amiodarone could be related. In previous report of animal models of acquired long QT syndrome, amiodarone increased QTc time in 6 of 7 dogs, while dispersion of repolarization was increased in 3. The three dogs tended to have higher tissue levels of amiodarone and its metabolite compared with those without dispersion of repolarization (van Opstal et al., 2001). In this case, we did not check the concentration of amiodarone or n‐desethylamiodarone; however, there was a possibility that they were quite high. 3 CONCLUSION Although the reported incidence of TdP during amiodarone therapy is low, careful ECG monitoring should be undergone to check not only QT interval but Tp‐e interval. CONFLICT OF INTEREST None. ETHICAL APPROVAL The authors have obtained the patient's informed consent. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
Recovered
ReactionOutcome
CC BY
33070441
18,469,623
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Pancreatitis'.
Red yeast rice (Monascus purpureus) supplements: Case series assessment of spontaneously reported cases to The Netherlands Pharmacovigilance Centre Lareb. Serious concerns are expressed on the safe use of red yeast rice (RYR) supplements. The aim of the present study was to analyse cases received by Lareb on RYR-related adverse health events. These cases were analysed for the number of reports, number of adverse drug reactions (ADRs), causality, seriousness of the reaction, latency-period, age and sex of the patients, concomitant medication and type of reporter. A total of 94 individual reports were collected by Lareb, corresponding with 187 ADRs. The analysis showed most reported ADRs were labelled musculoskeletal and connective tissue disorders (n = 64), gastrointestinal disorders (n = 33) and general disorders and administration site conditions (n = 23). The use of RYR supplements should be considered as a significant safety concern. Exposure to monacolin K could lead to serious adverse effects. To fully assess the safety profile of RYR supplements, more research is necessary to compose a complete ADR profile of RYR supplements. What is already known about this subject Red yeast rice (RYR) supplements are used to lower serum cholesterol levels in patients with statin intolerance. RYR supplements contain a wide variety of monacolin compounds, of which monacolin K is the primary form being chemically identical to lovastatin. Serious concerns exist on the safety of RYR supplements, for which general toxicological knowledge on the potential health risks and involved mechanism is lacking. What this study adds The present study reveals numerous cases of adverse health effects by RYR and proposes potential toxicological mechanisms involved. Our study confirms that the use of RYR supplements should be considered as a significant safety concern. 1 INTRODUCTION Statins belong to the first‐line therapy in the reduction of elevated blood cholesterol levels and have been shown to reduce the rate of cardiovascular events and reduce all‐time mortality. 1 , 2 However, statins have been associated with muscle‐related adverse events that are most commonly reported as a major reason for statin intolerance 3 Several patients with statin intolerance seek alternative products to reduce elevated blood cholesterol levels. Additionally, even healthy consumers use alternative products to lower serum cholesterol. One of the most common alternative products are red yeast rice (RYR) supplements 3 RYR is produced by fermenting rice with Monascus porpureus, forming monacolin compounds 1 The primary monacolin compound present in the RYR supplements is monacolin K, which is chemically identical to the pharmaceutical lovastatin 4 Monacolin K inhibits HMG‐CoA reductase, an enzyme that catalyses the conversion of HMG‐CoA to mevalonate, a precursor of cholesterol. Through the inhibition of HMG‐CoA, monacolin K decreases the production rate of cholesterol 1 , 5 Several clinical trials have shown that RYR supplements are associated with the prevention of primary and secondary cardiovascular events 4 , 6 , 7 , 8 Besides these effects, also anticancer, anti‐inflammatory, antihypertensive and antidiabetic effects have been described 5 , 9 In contrast to these beneficial health effects, serious concerns exist on the safety of RYR supplements. The structural similarity with lovastatin implies that similar adverse drug reactions (ADRs) can be expected. Overall, general toxicological knowledge on the potential health risks of RYR are lacking. Identification of potential risks of RYR products is challenging because food supplements are not usually registered in pharmacovigilance databases where suspected ADRs can be collected. However, The Netherlands Pharmacovigilance Centre Lareb has received numerous spontaneous reports of ADRs by RYR supplements over recent years. The aim of this article is to comprehensively describe the cases received and assess the case series concerning the use of RYR and health complications. 2 METHODS 2.1 Description of cases In the Netherlands, the Pharmacovigilance Centre Lareb maintains the spontaneous reporting system for drugs, vaccines and natural products with a health claim, such as herbal drugs. By collecting and analysing reported ADRs, knowledge is generated on the occurrence of ADRs in daily practice. Data on reported ADRs was gathered from 1991 (establishment of the centre) until March 2020. The reports can originate from health care professionals or consumers and can be received directly or through pharmaceutical companies. All directly received reports in the Dutch spontaneous reporting system are individually triaged and assessed by trained assessors at Lareb. For the purpose of this study additional causality assessed was performed by the authors according to the World Health Organization causality model to determine the role of RYR in ADR occurrence 10 ADRs are coded with the Medical Dictionary for Regulatory Activities (MedDRA) 11 In the case of unregistered products, Lareb uses a custom built drug dictionary for coding. 12 The number of reports, number of ADRs, seriousness of the reaction 13 latency‐period, age and sex of the patient, concomitant medication, and type of reporter were described. The reports were grouped based on the MedDRA system organ class (SOC) and preferred term (PT). A line‐listing of all selected cases was made. 3 RESULTS 3.1 Characteristics of reports From December 2007 to March 2020, a total of 94 individual reports were collected by Lareb, of which only 16 were received before June 2017. In these reports, in total 187 ADRs were reported, coded as PTs according the standardized MedDRA system. Six reports were according the CIOMS criteria classified as serious: 3 times (acute) pancreatitis and 2 times symptoms of rhabdomyolyses such as chromaturia, abnormal urine odor, myalgia, muscle spasms and malaise were reported. One report concerned acute hepatic failure with jaundice where the liver transplantation was required. World Health Organization causality assessment revealed the role of RYR in to be certain (n = 2), probably/likely (n = 24), possible (n = 61) and unlikely (n = 7) in the reported cases. The majority of reports (n = 75) were submitted by consumers or other non‐health professionals. Nine cases were reported by pharmacists, 8 cases by a physician and 2 reports were reported by both pharmacist and physician. Reported sex was 60 females and 34 males and the mean age was 64 years (range 17–81 years), the median age was 65 years. Concomitant medication use was reported in 55 reports (59.1%). The most reported comedication was vitamin D (colecalciferol), reported 11 times, followed by clopidogrel (Table S1). It must be noted that 1 report can contain multiple concomitant drugs. 3.2 Action and outcome In 66 reports, patients discontinued their treatment with RYR supplements; in 47 cases of these the symptoms resolved, were recovering or recovered with sequel. The following actions with RYR supplement occurred in the remaining patients: no adjustment (n = 13), unknown (n = 5), dose reduced (n = 2), not applicable (n = 6) and not reported (n = 2). 3.3 SOC reports A total of 94 individual reports of ADRs, corresponding to 187 PTs were related to RYR supplement use (Table S3). The most frequently reported PTs were labelled under the SOC musculoskeletal and connective tissue disorders, gastrointestinal disorders, general disorders and administration site conditions and nervous system disorders (Table S2). In total 64 PTs were reported in the SOC musculoskeletal and connective tissue disorders, which included myalgia, muscle spasms, weakness rhabdomyolysis, muscle atrophy, limb discomfort, flank pain, osteoarthritis and tendonitis. Gastrointestinal disorders was reported 33 times, and included inter alia (i.a.) abdominal pain, abdominal discomfort, nausea, diarrhoea and pancreatitis. General disorders and administration site conditions was reported 23 times and included i.a. fatigue, malaise and peripheral oedema. Nervous system disorders, such as headache, dizziness and sleep disorders, were reported 16 times. All SOCs and related no. of ADRs can be found in Table S2. 3.4 Latency Notable the median latency time for all reported ADRs is 2.1 weeks and also median latency's for the symptoms classified within the 4 most reported SOCs show the same tendency, as it falls between 1.9 weeks for nervous system disorders, 2 weeks for general symptoms, 2.4 weeks for musculoskeletal disorders and 2.9 weeks for gastrointestinal disorders (Figure 1B). FIGURE 1 Median latency times for the symptoms in the cases reported to Lareb. GD: general disorders and administration site conditions; NSD: nervous system disorders; GID: gastrointestinal disorders; MCD: musculoskeletal and connective tissue disorders; SOCs: system organ classes 4 DISCUSSION Although RYR‐containing products have been available on the market as food supplements for several years and their efficacy has been proven, 4 , 6 , 7 , 8 serious concerns about their safety profile remain. The present study reveals the most reported ADRs due to the use of RYR supplements to Lareb. It can be noted that the profile of reported ADRs to RYR of the present study is in line with previous studies, 14 , 15 , 16 ] with musculoskeletal and gastrointestinal, as the most frequently reported adverse effects (AEs). Although similar studies have been published before, these supplements are being used on a large scale and new cases of AEs due to these supplements arise. Our case series adds to the existing case reports and contributes to increasing the awareness for the potential adverse health effects of RYR supplements. Overall, our results demonstrate that the safety profile of RYR supplements is similar to that of the synthetic statins, particularly lovastatin. Several uncertainties exist that complicate drawing a complete safety profile of RYR supplements. First of all, there is a wide variability in composition of RYR‐containing food supplements. The monacolins in RYR are used in multi‐ingredient botanical preparations containing multiple other components, which have not been fully evaluated individually or in combination. Moreover, it is evident that the ratio of monacolin K and its hydroxyl acid form monacolin KA differs widely in numerous RYR products. 17 Other than monacolin K, there is a lack of data on the bioactivity of the other components in RYR. Therefore, it is difficult to assess the toxicological mechanisms of this mixture of monacolins. Since RYR most abundantly consists of monacolin K, which is a structural homologue to lovastatin, the mechanisms of RYR‐induced adverse effects are expected to be similar to those known for statins. 4.1 Musculoskeletal disorders Skeletal muscle abnormalities are the most reported and clinically important side effects of RYR, as shown in Table S2. Smith et al. published a case report of a middle‐ aged man presenting with muscle weakness after receiving RYR supplements for 2 months. 18 After examining the patients, they found increased creatine kinase levels of 385 IU/L (normal range is 30–160 IU/L). Additionally, Venhuis et al. reported a similar case report of patients using RYR products. 19 The patients exhibited multiple AEs, including myalgia. Although the exact mechanisms behind the musculoskeletal ADRs are not known, numerous studies have been performed on identification of the mechanism. 20 , 21 Multiple proposed mechanisms include inhibition of coenzyme Q10 synthesis, isoprenoid depletion, decreased or altered sarcolemma membrane cholesterol, disturbed calcium metabolism or autoimmune phenomena 22 , 23 , 24 , 25 The most obvious explanation for RYR‐induced myopathy would be the inhibition of mevalonate, a precursor of coenzyme Q10, due to the HMG‐CoA inhibition. As a consequence of this inhibition, the mitochondrial respiratory chain may be impaired, and this may result in impaired energy production and ultimately cell death 26 This effect is better described for statins, as several studies have shown lower levels of coenzyme Q10 after the use of statins 27 , 28 4.2 Gastrointestinal related disorders The cases described by the present study show that gastrointestinal reactions are often associated with the use of RYR supplements. These symptoms, varying from dyspepsia to vomiting and abdominal pain, nausea and diarrhoea, are also mostly reported by other studies 6 Mazzanti et al. also found a high percentage of gastrointestinal disorders: 22% of all reported ADRs. In general, the reactions were not serious with the exception of 1 hospitalization 15 Although there is no specific explanation yet for these side effects of RYR supplements, gastrointestinal reactions are not uncommon reactions associated with the use of statins, in which mitochondrial dysfunction is suggested to play an important role 29 , 30 4.3 Hepatic disorders Besides musculoskeletal and gastrointestinal disorders, adverse effects associated with RYR that are repeatedly reported are elevated levels of hepatic enzymes, indicative for liver injury. This is also shown by a meta‐analysis on the efficacy and safety of RYR supplements that included a total of 13 randomized controlled trials 31 The analysis showed that the serum alanine transaminase and aspartate aminotransferase levels were significantly increased in RYR users. This might explain the 3 cases of liver injury identified by the present study. In addition to the 3 cases reported by our study, multiple other cases of RYR‐induced liver toxicity have been documented 14 , 15 Three individual cases of severe hepatitis due to RYR were published previously, highlighting that, despite the low incidence of liver injury, the use of RYR supplements might have serious health consequences 32 , 33 , 34 The exact mechanism by which RYR causes hepatotoxicity is not well understood yet. However, it is expected that liver injury by RYR supplements may be attributed to the presence of lovastatin, as liver injuries are also among the most reported side effects of statin users 1 , 35 , 36 , 37 Similar to the musculoskeletal adverse effects, mitochondrial dysfunction is proposed to play a major role in the observed liver injury by statins 38 , 39 4.4 Kidney damage In our case series, kidney damage due to the use of RYR supplements was reported 5 times. The induced kidney damage might be explained by the presence of the mycotoxin citrinin in a significant number of RYR supplement, although this is unfortunately not measured 40 Citrinin is known to have nephrotoxic properties and therefore RYR supplements containing citrinin is a cause for concern, since it can pose a serious health risk: both in vivo and in vitro studies showed that citrinin has potentially mutagenic and mutatoxic properties 41 , 42 , 43 , 44 Another possible explanation for the toxic effects on the kidneys can be found in the muscle damage by RYR supplements. This can result in apoptosis of muscle cells, causing release myoglobin into the blood stream. In the kidneys, myoglobin can be converted to ferrihaem, which is considered a nephrotoxic compound 45 4.5 Possible interactions between comedication and RYR supplements Comedication influencing the bioavailability of monacolin K could also increase the chance of adverse health events by RYR supplements. The present case series shows that approximately 60% of the reported cases concerned the concomitant use of RYR supplements and medicines (Table S1). The use of comedication can potentially interfere with the kinetics and dynamics of RYR. Statins are metabolized by cytochrome P450 3A4 enzymes, and the administration of RYR with cytochrome enzyme inhibitors can therefore increase the risk of side effects by increasing monacolin K plasma concentrations. Such pharmacokinetic interaction has been reported in a 28‐year‐old kidney transplantation female patient who used a preparation containing RYR and cyclosporine 46 This patient developed rhabdomyolysis several months after starting the use of RYR, after receiving a combination of drugs in her post‐transplant period. Another case report suggests that coadministration of RYR with esomeprazole may increase the plasma concentrations of monacolin K and thus the associated risk of myopathy. The proposed mechanism is competitive inhibition of intestinal P‐glycoprotein, resulting in decreased drug secretion into the intestinal lumen and increased drug bioavailability 47 Also, pharmacodynamic interactions may occur by the combined use of drugs and RYR supplements. This will especially be relevant in case of taking RYR together with other statins. In 2 of our cases, atorvastatin was reported as comedication. This means that a potentiation of the HMCG‐CoA inhibition may occur, potentially leading to an increased risk for the occurrence of the ADRs. Hence, the use of RYR in combination with other drugs needs to be done carefully due to potential interactions that may lead to serious adverse health effects. 5 LIMITATIONS In the assessment of case series of spontaneously reported cases, several limitations could play a role. Whereas comedications potentially cause interactions with RYR supplements and thereby increase the chance of developing ADRs, they may also have a confounding effect that complicates identifying the role of RYR in the ADRs. There are, for instance, concomitant drugs known to be potentially associated with pancreatitis, such as morphine, perindopril and telmisartan. To exclude this confounding effect of drugs as much as possible, the use of comedications is taken into account in the causality assessment. Another potential limitation of the present case series of spontaneous reports is notoriety bias 48 Although notoriety bias always plays a role when analysing and discussing spontaneous reports, this does not mean that the reported cases to Lareb are no real cases, only that the reporter was triggered to submit their case after the media attention. Seeing that the cases are reported over a period of multiple years, the increase in reports is likely to reflect the growing use of RYR products and is not merely based on more media attention. Finally, the absence of information about content of monacolin K makes it difficult to strongly relate the ADRs to the use of RYR supplements. However, even if such information were available, this would still provide uncertain information for 2 reasons: (i) based on the reports, it cannot be determined how many dosages 1 patient used; (ii) as the supplements were generally not chemically analysed, it remains uncertain what exactly, and how much, is present of monacolins in the supplement. Interestingly, the analysis of 1 RYR sample of 1 of the reports showed that the dose of monacolin K in the supplements varied between pills from 1 jar (7 vs 3 mg). It is therefore impossible to assess the chemicals and doses to which the patients were exposed. 6 CONCLUSION The intake of monacolins via dietary supplements can lead to serious adverse health effects. Monacolin K is chemically identical to lovastatin and therefore these adverse health effects are similar to those of lovastatin. The use of RYR supplements should therefore be considered as a significant safety concern. So far, exact mechanisms on these toxic effects have not been established. This can be explained by the fact that the RYR supplements contain a wide variety of chemical compounds, most of which have not been toxicologically characterized. To fully assess the safety profile of RYR supplements, more research is needed into all these chemical constituents. Based on remaining uncertainties on the effects of RYR supplements, it remains impossible to identify recommended dietary intake levels of monacolins from RYR that would not give any harmful effect to human health. COMPETING INTERESTS There are no competing interests to declare. CONTRIBUTORS All authors substantially contributed to designing the study, acquiring, analyzing and interpreting the data; and drafting and revising the work. All authors approved the submitted final version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. Supporting information TABLE S1 Top 10 reported comedications Click here for additional data file. TABLE S2 Number of reported ADRs per SOC Click here for additional data file. TABLE S3 Overview of all reported cases to Lareb. Click here for additional data file. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
ACETAMINOPHEN, AMITRIPTYLINE, CARBASPIRIN CALCIUM, CHOLECALCIFEROL, DIETARY SUPPLEMENT, METOCLOPRAMIDE, METOPROLOL, MORPHINE, NADROPARIN, OXAZEPAM, PANTOPRAZOLE
DrugsGivenReaction
CC BY-NC
33085778
19,715,798
2021-04
What was the outcome of reaction 'Cholecystitis'?
Red yeast rice (Monascus purpureus) supplements: Case series assessment of spontaneously reported cases to The Netherlands Pharmacovigilance Centre Lareb. Serious concerns are expressed on the safe use of red yeast rice (RYR) supplements. The aim of the present study was to analyse cases received by Lareb on RYR-related adverse health events. These cases were analysed for the number of reports, number of adverse drug reactions (ADRs), causality, seriousness of the reaction, latency-period, age and sex of the patients, concomitant medication and type of reporter. A total of 94 individual reports were collected by Lareb, corresponding with 187 ADRs. The analysis showed most reported ADRs were labelled musculoskeletal and connective tissue disorders (n = 64), gastrointestinal disorders (n = 33) and general disorders and administration site conditions (n = 23). The use of RYR supplements should be considered as a significant safety concern. Exposure to monacolin K could lead to serious adverse effects. To fully assess the safety profile of RYR supplements, more research is necessary to compose a complete ADR profile of RYR supplements. What is already known about this subject Red yeast rice (RYR) supplements are used to lower serum cholesterol levels in patients with statin intolerance. RYR supplements contain a wide variety of monacolin compounds, of which monacolin K is the primary form being chemically identical to lovastatin. Serious concerns exist on the safety of RYR supplements, for which general toxicological knowledge on the potential health risks and involved mechanism is lacking. What this study adds The present study reveals numerous cases of adverse health effects by RYR and proposes potential toxicological mechanisms involved. Our study confirms that the use of RYR supplements should be considered as a significant safety concern. 1 INTRODUCTION Statins belong to the first‐line therapy in the reduction of elevated blood cholesterol levels and have been shown to reduce the rate of cardiovascular events and reduce all‐time mortality. 1 , 2 However, statins have been associated with muscle‐related adverse events that are most commonly reported as a major reason for statin intolerance 3 Several patients with statin intolerance seek alternative products to reduce elevated blood cholesterol levels. Additionally, even healthy consumers use alternative products to lower serum cholesterol. One of the most common alternative products are red yeast rice (RYR) supplements 3 RYR is produced by fermenting rice with Monascus porpureus, forming monacolin compounds 1 The primary monacolin compound present in the RYR supplements is monacolin K, which is chemically identical to the pharmaceutical lovastatin 4 Monacolin K inhibits HMG‐CoA reductase, an enzyme that catalyses the conversion of HMG‐CoA to mevalonate, a precursor of cholesterol. Through the inhibition of HMG‐CoA, monacolin K decreases the production rate of cholesterol 1 , 5 Several clinical trials have shown that RYR supplements are associated with the prevention of primary and secondary cardiovascular events 4 , 6 , 7 , 8 Besides these effects, also anticancer, anti‐inflammatory, antihypertensive and antidiabetic effects have been described 5 , 9 In contrast to these beneficial health effects, serious concerns exist on the safety of RYR supplements. The structural similarity with lovastatin implies that similar adverse drug reactions (ADRs) can be expected. Overall, general toxicological knowledge on the potential health risks of RYR are lacking. Identification of potential risks of RYR products is challenging because food supplements are not usually registered in pharmacovigilance databases where suspected ADRs can be collected. However, The Netherlands Pharmacovigilance Centre Lareb has received numerous spontaneous reports of ADRs by RYR supplements over recent years. The aim of this article is to comprehensively describe the cases received and assess the case series concerning the use of RYR and health complications. 2 METHODS 2.1 Description of cases In the Netherlands, the Pharmacovigilance Centre Lareb maintains the spontaneous reporting system for drugs, vaccines and natural products with a health claim, such as herbal drugs. By collecting and analysing reported ADRs, knowledge is generated on the occurrence of ADRs in daily practice. Data on reported ADRs was gathered from 1991 (establishment of the centre) until March 2020. The reports can originate from health care professionals or consumers and can be received directly or through pharmaceutical companies. All directly received reports in the Dutch spontaneous reporting system are individually triaged and assessed by trained assessors at Lareb. For the purpose of this study additional causality assessed was performed by the authors according to the World Health Organization causality model to determine the role of RYR in ADR occurrence 10 ADRs are coded with the Medical Dictionary for Regulatory Activities (MedDRA) 11 In the case of unregistered products, Lareb uses a custom built drug dictionary for coding. 12 The number of reports, number of ADRs, seriousness of the reaction 13 latency‐period, age and sex of the patient, concomitant medication, and type of reporter were described. The reports were grouped based on the MedDRA system organ class (SOC) and preferred term (PT). A line‐listing of all selected cases was made. 3 RESULTS 3.1 Characteristics of reports From December 2007 to March 2020, a total of 94 individual reports were collected by Lareb, of which only 16 were received before June 2017. In these reports, in total 187 ADRs were reported, coded as PTs according the standardized MedDRA system. Six reports were according the CIOMS criteria classified as serious: 3 times (acute) pancreatitis and 2 times symptoms of rhabdomyolyses such as chromaturia, abnormal urine odor, myalgia, muscle spasms and malaise were reported. One report concerned acute hepatic failure with jaundice where the liver transplantation was required. World Health Organization causality assessment revealed the role of RYR in to be certain (n = 2), probably/likely (n = 24), possible (n = 61) and unlikely (n = 7) in the reported cases. The majority of reports (n = 75) were submitted by consumers or other non‐health professionals. Nine cases were reported by pharmacists, 8 cases by a physician and 2 reports were reported by both pharmacist and physician. Reported sex was 60 females and 34 males and the mean age was 64 years (range 17–81 years), the median age was 65 years. Concomitant medication use was reported in 55 reports (59.1%). The most reported comedication was vitamin D (colecalciferol), reported 11 times, followed by clopidogrel (Table S1). It must be noted that 1 report can contain multiple concomitant drugs. 3.2 Action and outcome In 66 reports, patients discontinued their treatment with RYR supplements; in 47 cases of these the symptoms resolved, were recovering or recovered with sequel. The following actions with RYR supplement occurred in the remaining patients: no adjustment (n = 13), unknown (n = 5), dose reduced (n = 2), not applicable (n = 6) and not reported (n = 2). 3.3 SOC reports A total of 94 individual reports of ADRs, corresponding to 187 PTs were related to RYR supplement use (Table S3). The most frequently reported PTs were labelled under the SOC musculoskeletal and connective tissue disorders, gastrointestinal disorders, general disorders and administration site conditions and nervous system disorders (Table S2). In total 64 PTs were reported in the SOC musculoskeletal and connective tissue disorders, which included myalgia, muscle spasms, weakness rhabdomyolysis, muscle atrophy, limb discomfort, flank pain, osteoarthritis and tendonitis. Gastrointestinal disorders was reported 33 times, and included inter alia (i.a.) abdominal pain, abdominal discomfort, nausea, diarrhoea and pancreatitis. General disorders and administration site conditions was reported 23 times and included i.a. fatigue, malaise and peripheral oedema. Nervous system disorders, such as headache, dizziness and sleep disorders, were reported 16 times. All SOCs and related no. of ADRs can be found in Table S2. 3.4 Latency Notable the median latency time for all reported ADRs is 2.1 weeks and also median latency's for the symptoms classified within the 4 most reported SOCs show the same tendency, as it falls between 1.9 weeks for nervous system disorders, 2 weeks for general symptoms, 2.4 weeks for musculoskeletal disorders and 2.9 weeks for gastrointestinal disorders (Figure 1B). FIGURE 1 Median latency times for the symptoms in the cases reported to Lareb. GD: general disorders and administration site conditions; NSD: nervous system disorders; GID: gastrointestinal disorders; MCD: musculoskeletal and connective tissue disorders; SOCs: system organ classes 4 DISCUSSION Although RYR‐containing products have been available on the market as food supplements for several years and their efficacy has been proven, 4 , 6 , 7 , 8 serious concerns about their safety profile remain. The present study reveals the most reported ADRs due to the use of RYR supplements to Lareb. It can be noted that the profile of reported ADRs to RYR of the present study is in line with previous studies, 14 , 15 , 16 ] with musculoskeletal and gastrointestinal, as the most frequently reported adverse effects (AEs). Although similar studies have been published before, these supplements are being used on a large scale and new cases of AEs due to these supplements arise. Our case series adds to the existing case reports and contributes to increasing the awareness for the potential adverse health effects of RYR supplements. Overall, our results demonstrate that the safety profile of RYR supplements is similar to that of the synthetic statins, particularly lovastatin. Several uncertainties exist that complicate drawing a complete safety profile of RYR supplements. First of all, there is a wide variability in composition of RYR‐containing food supplements. The monacolins in RYR are used in multi‐ingredient botanical preparations containing multiple other components, which have not been fully evaluated individually or in combination. Moreover, it is evident that the ratio of monacolin K and its hydroxyl acid form monacolin KA differs widely in numerous RYR products. 17 Other than monacolin K, there is a lack of data on the bioactivity of the other components in RYR. Therefore, it is difficult to assess the toxicological mechanisms of this mixture of monacolins. Since RYR most abundantly consists of monacolin K, which is a structural homologue to lovastatin, the mechanisms of RYR‐induced adverse effects are expected to be similar to those known for statins. 4.1 Musculoskeletal disorders Skeletal muscle abnormalities are the most reported and clinically important side effects of RYR, as shown in Table S2. Smith et al. published a case report of a middle‐ aged man presenting with muscle weakness after receiving RYR supplements for 2 months. 18 After examining the patients, they found increased creatine kinase levels of 385 IU/L (normal range is 30–160 IU/L). Additionally, Venhuis et al. reported a similar case report of patients using RYR products. 19 The patients exhibited multiple AEs, including myalgia. Although the exact mechanisms behind the musculoskeletal ADRs are not known, numerous studies have been performed on identification of the mechanism. 20 , 21 Multiple proposed mechanisms include inhibition of coenzyme Q10 synthesis, isoprenoid depletion, decreased or altered sarcolemma membrane cholesterol, disturbed calcium metabolism or autoimmune phenomena 22 , 23 , 24 , 25 The most obvious explanation for RYR‐induced myopathy would be the inhibition of mevalonate, a precursor of coenzyme Q10, due to the HMG‐CoA inhibition. As a consequence of this inhibition, the mitochondrial respiratory chain may be impaired, and this may result in impaired energy production and ultimately cell death 26 This effect is better described for statins, as several studies have shown lower levels of coenzyme Q10 after the use of statins 27 , 28 4.2 Gastrointestinal related disorders The cases described by the present study show that gastrointestinal reactions are often associated with the use of RYR supplements. These symptoms, varying from dyspepsia to vomiting and abdominal pain, nausea and diarrhoea, are also mostly reported by other studies 6 Mazzanti et al. also found a high percentage of gastrointestinal disorders: 22% of all reported ADRs. In general, the reactions were not serious with the exception of 1 hospitalization 15 Although there is no specific explanation yet for these side effects of RYR supplements, gastrointestinal reactions are not uncommon reactions associated with the use of statins, in which mitochondrial dysfunction is suggested to play an important role 29 , 30 4.3 Hepatic disorders Besides musculoskeletal and gastrointestinal disorders, adverse effects associated with RYR that are repeatedly reported are elevated levels of hepatic enzymes, indicative for liver injury. This is also shown by a meta‐analysis on the efficacy and safety of RYR supplements that included a total of 13 randomized controlled trials 31 The analysis showed that the serum alanine transaminase and aspartate aminotransferase levels were significantly increased in RYR users. This might explain the 3 cases of liver injury identified by the present study. In addition to the 3 cases reported by our study, multiple other cases of RYR‐induced liver toxicity have been documented 14 , 15 Three individual cases of severe hepatitis due to RYR were published previously, highlighting that, despite the low incidence of liver injury, the use of RYR supplements might have serious health consequences 32 , 33 , 34 The exact mechanism by which RYR causes hepatotoxicity is not well understood yet. However, it is expected that liver injury by RYR supplements may be attributed to the presence of lovastatin, as liver injuries are also among the most reported side effects of statin users 1 , 35 , 36 , 37 Similar to the musculoskeletal adverse effects, mitochondrial dysfunction is proposed to play a major role in the observed liver injury by statins 38 , 39 4.4 Kidney damage In our case series, kidney damage due to the use of RYR supplements was reported 5 times. The induced kidney damage might be explained by the presence of the mycotoxin citrinin in a significant number of RYR supplement, although this is unfortunately not measured 40 Citrinin is known to have nephrotoxic properties and therefore RYR supplements containing citrinin is a cause for concern, since it can pose a serious health risk: both in vivo and in vitro studies showed that citrinin has potentially mutagenic and mutatoxic properties 41 , 42 , 43 , 44 Another possible explanation for the toxic effects on the kidneys can be found in the muscle damage by RYR supplements. This can result in apoptosis of muscle cells, causing release myoglobin into the blood stream. In the kidneys, myoglobin can be converted to ferrihaem, which is considered a nephrotoxic compound 45 4.5 Possible interactions between comedication and RYR supplements Comedication influencing the bioavailability of monacolin K could also increase the chance of adverse health events by RYR supplements. The present case series shows that approximately 60% of the reported cases concerned the concomitant use of RYR supplements and medicines (Table S1). The use of comedication can potentially interfere with the kinetics and dynamics of RYR. Statins are metabolized by cytochrome P450 3A4 enzymes, and the administration of RYR with cytochrome enzyme inhibitors can therefore increase the risk of side effects by increasing monacolin K plasma concentrations. Such pharmacokinetic interaction has been reported in a 28‐year‐old kidney transplantation female patient who used a preparation containing RYR and cyclosporine 46 This patient developed rhabdomyolysis several months after starting the use of RYR, after receiving a combination of drugs in her post‐transplant period. Another case report suggests that coadministration of RYR with esomeprazole may increase the plasma concentrations of monacolin K and thus the associated risk of myopathy. The proposed mechanism is competitive inhibition of intestinal P‐glycoprotein, resulting in decreased drug secretion into the intestinal lumen and increased drug bioavailability 47 Also, pharmacodynamic interactions may occur by the combined use of drugs and RYR supplements. This will especially be relevant in case of taking RYR together with other statins. In 2 of our cases, atorvastatin was reported as comedication. This means that a potentiation of the HMCG‐CoA inhibition may occur, potentially leading to an increased risk for the occurrence of the ADRs. Hence, the use of RYR in combination with other drugs needs to be done carefully due to potential interactions that may lead to serious adverse health effects. 5 LIMITATIONS In the assessment of case series of spontaneously reported cases, several limitations could play a role. Whereas comedications potentially cause interactions with RYR supplements and thereby increase the chance of developing ADRs, they may also have a confounding effect that complicates identifying the role of RYR in the ADRs. There are, for instance, concomitant drugs known to be potentially associated with pancreatitis, such as morphine, perindopril and telmisartan. To exclude this confounding effect of drugs as much as possible, the use of comedications is taken into account in the causality assessment. Another potential limitation of the present case series of spontaneous reports is notoriety bias 48 Although notoriety bias always plays a role when analysing and discussing spontaneous reports, this does not mean that the reported cases to Lareb are no real cases, only that the reporter was triggered to submit their case after the media attention. Seeing that the cases are reported over a period of multiple years, the increase in reports is likely to reflect the growing use of RYR products and is not merely based on more media attention. Finally, the absence of information about content of monacolin K makes it difficult to strongly relate the ADRs to the use of RYR supplements. However, even if such information were available, this would still provide uncertain information for 2 reasons: (i) based on the reports, it cannot be determined how many dosages 1 patient used; (ii) as the supplements were generally not chemically analysed, it remains uncertain what exactly, and how much, is present of monacolins in the supplement. Interestingly, the analysis of 1 RYR sample of 1 of the reports showed that the dose of monacolin K in the supplements varied between pills from 1 jar (7 vs 3 mg). It is therefore impossible to assess the chemicals and doses to which the patients were exposed. 6 CONCLUSION The intake of monacolins via dietary supplements can lead to serious adverse health effects. Monacolin K is chemically identical to lovastatin and therefore these adverse health effects are similar to those of lovastatin. The use of RYR supplements should therefore be considered as a significant safety concern. So far, exact mechanisms on these toxic effects have not been established. This can be explained by the fact that the RYR supplements contain a wide variety of chemical compounds, most of which have not been toxicologically characterized. To fully assess the safety profile of RYR supplements, more research is needed into all these chemical constituents. Based on remaining uncertainties on the effects of RYR supplements, it remains impossible to identify recommended dietary intake levels of monacolins from RYR that would not give any harmful effect to human health. COMPETING INTERESTS There are no competing interests to declare. CONTRIBUTORS All authors substantially contributed to designing the study, acquiring, analyzing and interpreting the data; and drafting and revising the work. All authors approved the submitted final version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. Supporting information TABLE S1 Top 10 reported comedications Click here for additional data file. TABLE S2 Number of reported ADRs per SOC Click here for additional data file. TABLE S3 Overview of all reported cases to Lareb. Click here for additional data file. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
Recovering
ReactionOutcome
CC BY-NC
33085778
19,715,717
2021-04
What was the outcome of reaction 'Pancreatitis acute'?
Red yeast rice (Monascus purpureus) supplements: Case series assessment of spontaneously reported cases to The Netherlands Pharmacovigilance Centre Lareb. Serious concerns are expressed on the safe use of red yeast rice (RYR) supplements. The aim of the present study was to analyse cases received by Lareb on RYR-related adverse health events. These cases were analysed for the number of reports, number of adverse drug reactions (ADRs), causality, seriousness of the reaction, latency-period, age and sex of the patients, concomitant medication and type of reporter. A total of 94 individual reports were collected by Lareb, corresponding with 187 ADRs. The analysis showed most reported ADRs were labelled musculoskeletal and connective tissue disorders (n = 64), gastrointestinal disorders (n = 33) and general disorders and administration site conditions (n = 23). The use of RYR supplements should be considered as a significant safety concern. Exposure to monacolin K could lead to serious adverse effects. To fully assess the safety profile of RYR supplements, more research is necessary to compose a complete ADR profile of RYR supplements. What is already known about this subject Red yeast rice (RYR) supplements are used to lower serum cholesterol levels in patients with statin intolerance. RYR supplements contain a wide variety of monacolin compounds, of which monacolin K is the primary form being chemically identical to lovastatin. Serious concerns exist on the safety of RYR supplements, for which general toxicological knowledge on the potential health risks and involved mechanism is lacking. What this study adds The present study reveals numerous cases of adverse health effects by RYR and proposes potential toxicological mechanisms involved. Our study confirms that the use of RYR supplements should be considered as a significant safety concern. 1 INTRODUCTION Statins belong to the first‐line therapy in the reduction of elevated blood cholesterol levels and have been shown to reduce the rate of cardiovascular events and reduce all‐time mortality. 1 , 2 However, statins have been associated with muscle‐related adverse events that are most commonly reported as a major reason for statin intolerance 3 Several patients with statin intolerance seek alternative products to reduce elevated blood cholesterol levels. Additionally, even healthy consumers use alternative products to lower serum cholesterol. One of the most common alternative products are red yeast rice (RYR) supplements 3 RYR is produced by fermenting rice with Monascus porpureus, forming monacolin compounds 1 The primary monacolin compound present in the RYR supplements is monacolin K, which is chemically identical to the pharmaceutical lovastatin 4 Monacolin K inhibits HMG‐CoA reductase, an enzyme that catalyses the conversion of HMG‐CoA to mevalonate, a precursor of cholesterol. Through the inhibition of HMG‐CoA, monacolin K decreases the production rate of cholesterol 1 , 5 Several clinical trials have shown that RYR supplements are associated with the prevention of primary and secondary cardiovascular events 4 , 6 , 7 , 8 Besides these effects, also anticancer, anti‐inflammatory, antihypertensive and antidiabetic effects have been described 5 , 9 In contrast to these beneficial health effects, serious concerns exist on the safety of RYR supplements. The structural similarity with lovastatin implies that similar adverse drug reactions (ADRs) can be expected. Overall, general toxicological knowledge on the potential health risks of RYR are lacking. Identification of potential risks of RYR products is challenging because food supplements are not usually registered in pharmacovigilance databases where suspected ADRs can be collected. However, The Netherlands Pharmacovigilance Centre Lareb has received numerous spontaneous reports of ADRs by RYR supplements over recent years. The aim of this article is to comprehensively describe the cases received and assess the case series concerning the use of RYR and health complications. 2 METHODS 2.1 Description of cases In the Netherlands, the Pharmacovigilance Centre Lareb maintains the spontaneous reporting system for drugs, vaccines and natural products with a health claim, such as herbal drugs. By collecting and analysing reported ADRs, knowledge is generated on the occurrence of ADRs in daily practice. Data on reported ADRs was gathered from 1991 (establishment of the centre) until March 2020. The reports can originate from health care professionals or consumers and can be received directly or through pharmaceutical companies. All directly received reports in the Dutch spontaneous reporting system are individually triaged and assessed by trained assessors at Lareb. For the purpose of this study additional causality assessed was performed by the authors according to the World Health Organization causality model to determine the role of RYR in ADR occurrence 10 ADRs are coded with the Medical Dictionary for Regulatory Activities (MedDRA) 11 In the case of unregistered products, Lareb uses a custom built drug dictionary for coding. 12 The number of reports, number of ADRs, seriousness of the reaction 13 latency‐period, age and sex of the patient, concomitant medication, and type of reporter were described. The reports were grouped based on the MedDRA system organ class (SOC) and preferred term (PT). A line‐listing of all selected cases was made. 3 RESULTS 3.1 Characteristics of reports From December 2007 to March 2020, a total of 94 individual reports were collected by Lareb, of which only 16 were received before June 2017. In these reports, in total 187 ADRs were reported, coded as PTs according the standardized MedDRA system. Six reports were according the CIOMS criteria classified as serious: 3 times (acute) pancreatitis and 2 times symptoms of rhabdomyolyses such as chromaturia, abnormal urine odor, myalgia, muscle spasms and malaise were reported. One report concerned acute hepatic failure with jaundice where the liver transplantation was required. World Health Organization causality assessment revealed the role of RYR in to be certain (n = 2), probably/likely (n = 24), possible (n = 61) and unlikely (n = 7) in the reported cases. The majority of reports (n = 75) were submitted by consumers or other non‐health professionals. Nine cases were reported by pharmacists, 8 cases by a physician and 2 reports were reported by both pharmacist and physician. Reported sex was 60 females and 34 males and the mean age was 64 years (range 17–81 years), the median age was 65 years. Concomitant medication use was reported in 55 reports (59.1%). The most reported comedication was vitamin D (colecalciferol), reported 11 times, followed by clopidogrel (Table S1). It must be noted that 1 report can contain multiple concomitant drugs. 3.2 Action and outcome In 66 reports, patients discontinued their treatment with RYR supplements; in 47 cases of these the symptoms resolved, were recovering or recovered with sequel. The following actions with RYR supplement occurred in the remaining patients: no adjustment (n = 13), unknown (n = 5), dose reduced (n = 2), not applicable (n = 6) and not reported (n = 2). 3.3 SOC reports A total of 94 individual reports of ADRs, corresponding to 187 PTs were related to RYR supplement use (Table S3). The most frequently reported PTs were labelled under the SOC musculoskeletal and connective tissue disorders, gastrointestinal disorders, general disorders and administration site conditions and nervous system disorders (Table S2). In total 64 PTs were reported in the SOC musculoskeletal and connective tissue disorders, which included myalgia, muscle spasms, weakness rhabdomyolysis, muscle atrophy, limb discomfort, flank pain, osteoarthritis and tendonitis. Gastrointestinal disorders was reported 33 times, and included inter alia (i.a.) abdominal pain, abdominal discomfort, nausea, diarrhoea and pancreatitis. General disorders and administration site conditions was reported 23 times and included i.a. fatigue, malaise and peripheral oedema. Nervous system disorders, such as headache, dizziness and sleep disorders, were reported 16 times. All SOCs and related no. of ADRs can be found in Table S2. 3.4 Latency Notable the median latency time for all reported ADRs is 2.1 weeks and also median latency's for the symptoms classified within the 4 most reported SOCs show the same tendency, as it falls between 1.9 weeks for nervous system disorders, 2 weeks for general symptoms, 2.4 weeks for musculoskeletal disorders and 2.9 weeks for gastrointestinal disorders (Figure 1B). FIGURE 1 Median latency times for the symptoms in the cases reported to Lareb. GD: general disorders and administration site conditions; NSD: nervous system disorders; GID: gastrointestinal disorders; MCD: musculoskeletal and connective tissue disorders; SOCs: system organ classes 4 DISCUSSION Although RYR‐containing products have been available on the market as food supplements for several years and their efficacy has been proven, 4 , 6 , 7 , 8 serious concerns about their safety profile remain. The present study reveals the most reported ADRs due to the use of RYR supplements to Lareb. It can be noted that the profile of reported ADRs to RYR of the present study is in line with previous studies, 14 , 15 , 16 ] with musculoskeletal and gastrointestinal, as the most frequently reported adverse effects (AEs). Although similar studies have been published before, these supplements are being used on a large scale and new cases of AEs due to these supplements arise. Our case series adds to the existing case reports and contributes to increasing the awareness for the potential adverse health effects of RYR supplements. Overall, our results demonstrate that the safety profile of RYR supplements is similar to that of the synthetic statins, particularly lovastatin. Several uncertainties exist that complicate drawing a complete safety profile of RYR supplements. First of all, there is a wide variability in composition of RYR‐containing food supplements. The monacolins in RYR are used in multi‐ingredient botanical preparations containing multiple other components, which have not been fully evaluated individually or in combination. Moreover, it is evident that the ratio of monacolin K and its hydroxyl acid form monacolin KA differs widely in numerous RYR products. 17 Other than monacolin K, there is a lack of data on the bioactivity of the other components in RYR. Therefore, it is difficult to assess the toxicological mechanisms of this mixture of monacolins. Since RYR most abundantly consists of monacolin K, which is a structural homologue to lovastatin, the mechanisms of RYR‐induced adverse effects are expected to be similar to those known for statins. 4.1 Musculoskeletal disorders Skeletal muscle abnormalities are the most reported and clinically important side effects of RYR, as shown in Table S2. Smith et al. published a case report of a middle‐ aged man presenting with muscle weakness after receiving RYR supplements for 2 months. 18 After examining the patients, they found increased creatine kinase levels of 385 IU/L (normal range is 30–160 IU/L). Additionally, Venhuis et al. reported a similar case report of patients using RYR products. 19 The patients exhibited multiple AEs, including myalgia. Although the exact mechanisms behind the musculoskeletal ADRs are not known, numerous studies have been performed on identification of the mechanism. 20 , 21 Multiple proposed mechanisms include inhibition of coenzyme Q10 synthesis, isoprenoid depletion, decreased or altered sarcolemma membrane cholesterol, disturbed calcium metabolism or autoimmune phenomena 22 , 23 , 24 , 25 The most obvious explanation for RYR‐induced myopathy would be the inhibition of mevalonate, a precursor of coenzyme Q10, due to the HMG‐CoA inhibition. As a consequence of this inhibition, the mitochondrial respiratory chain may be impaired, and this may result in impaired energy production and ultimately cell death 26 This effect is better described for statins, as several studies have shown lower levels of coenzyme Q10 after the use of statins 27 , 28 4.2 Gastrointestinal related disorders The cases described by the present study show that gastrointestinal reactions are often associated with the use of RYR supplements. These symptoms, varying from dyspepsia to vomiting and abdominal pain, nausea and diarrhoea, are also mostly reported by other studies 6 Mazzanti et al. also found a high percentage of gastrointestinal disorders: 22% of all reported ADRs. In general, the reactions were not serious with the exception of 1 hospitalization 15 Although there is no specific explanation yet for these side effects of RYR supplements, gastrointestinal reactions are not uncommon reactions associated with the use of statins, in which mitochondrial dysfunction is suggested to play an important role 29 , 30 4.3 Hepatic disorders Besides musculoskeletal and gastrointestinal disorders, adverse effects associated with RYR that are repeatedly reported are elevated levels of hepatic enzymes, indicative for liver injury. This is also shown by a meta‐analysis on the efficacy and safety of RYR supplements that included a total of 13 randomized controlled trials 31 The analysis showed that the serum alanine transaminase and aspartate aminotransferase levels were significantly increased in RYR users. This might explain the 3 cases of liver injury identified by the present study. In addition to the 3 cases reported by our study, multiple other cases of RYR‐induced liver toxicity have been documented 14 , 15 Three individual cases of severe hepatitis due to RYR were published previously, highlighting that, despite the low incidence of liver injury, the use of RYR supplements might have serious health consequences 32 , 33 , 34 The exact mechanism by which RYR causes hepatotoxicity is not well understood yet. However, it is expected that liver injury by RYR supplements may be attributed to the presence of lovastatin, as liver injuries are also among the most reported side effects of statin users 1 , 35 , 36 , 37 Similar to the musculoskeletal adverse effects, mitochondrial dysfunction is proposed to play a major role in the observed liver injury by statins 38 , 39 4.4 Kidney damage In our case series, kidney damage due to the use of RYR supplements was reported 5 times. The induced kidney damage might be explained by the presence of the mycotoxin citrinin in a significant number of RYR supplement, although this is unfortunately not measured 40 Citrinin is known to have nephrotoxic properties and therefore RYR supplements containing citrinin is a cause for concern, since it can pose a serious health risk: both in vivo and in vitro studies showed that citrinin has potentially mutagenic and mutatoxic properties 41 , 42 , 43 , 44 Another possible explanation for the toxic effects on the kidneys can be found in the muscle damage by RYR supplements. This can result in apoptosis of muscle cells, causing release myoglobin into the blood stream. In the kidneys, myoglobin can be converted to ferrihaem, which is considered a nephrotoxic compound 45 4.5 Possible interactions between comedication and RYR supplements Comedication influencing the bioavailability of monacolin K could also increase the chance of adverse health events by RYR supplements. The present case series shows that approximately 60% of the reported cases concerned the concomitant use of RYR supplements and medicines (Table S1). The use of comedication can potentially interfere with the kinetics and dynamics of RYR. Statins are metabolized by cytochrome P450 3A4 enzymes, and the administration of RYR with cytochrome enzyme inhibitors can therefore increase the risk of side effects by increasing monacolin K plasma concentrations. Such pharmacokinetic interaction has been reported in a 28‐year‐old kidney transplantation female patient who used a preparation containing RYR and cyclosporine 46 This patient developed rhabdomyolysis several months after starting the use of RYR, after receiving a combination of drugs in her post‐transplant period. Another case report suggests that coadministration of RYR with esomeprazole may increase the plasma concentrations of monacolin K and thus the associated risk of myopathy. The proposed mechanism is competitive inhibition of intestinal P‐glycoprotein, resulting in decreased drug secretion into the intestinal lumen and increased drug bioavailability 47 Also, pharmacodynamic interactions may occur by the combined use of drugs and RYR supplements. This will especially be relevant in case of taking RYR together with other statins. In 2 of our cases, atorvastatin was reported as comedication. This means that a potentiation of the HMCG‐CoA inhibition may occur, potentially leading to an increased risk for the occurrence of the ADRs. Hence, the use of RYR in combination with other drugs needs to be done carefully due to potential interactions that may lead to serious adverse health effects. 5 LIMITATIONS In the assessment of case series of spontaneously reported cases, several limitations could play a role. Whereas comedications potentially cause interactions with RYR supplements and thereby increase the chance of developing ADRs, they may also have a confounding effect that complicates identifying the role of RYR in the ADRs. There are, for instance, concomitant drugs known to be potentially associated with pancreatitis, such as morphine, perindopril and telmisartan. To exclude this confounding effect of drugs as much as possible, the use of comedications is taken into account in the causality assessment. Another potential limitation of the present case series of spontaneous reports is notoriety bias 48 Although notoriety bias always plays a role when analysing and discussing spontaneous reports, this does not mean that the reported cases to Lareb are no real cases, only that the reporter was triggered to submit their case after the media attention. Seeing that the cases are reported over a period of multiple years, the increase in reports is likely to reflect the growing use of RYR products and is not merely based on more media attention. Finally, the absence of information about content of monacolin K makes it difficult to strongly relate the ADRs to the use of RYR supplements. However, even if such information were available, this would still provide uncertain information for 2 reasons: (i) based on the reports, it cannot be determined how many dosages 1 patient used; (ii) as the supplements were generally not chemically analysed, it remains uncertain what exactly, and how much, is present of monacolins in the supplement. Interestingly, the analysis of 1 RYR sample of 1 of the reports showed that the dose of monacolin K in the supplements varied between pills from 1 jar (7 vs 3 mg). It is therefore impossible to assess the chemicals and doses to which the patients were exposed. 6 CONCLUSION The intake of monacolins via dietary supplements can lead to serious adverse health effects. Monacolin K is chemically identical to lovastatin and therefore these adverse health effects are similar to those of lovastatin. The use of RYR supplements should therefore be considered as a significant safety concern. So far, exact mechanisms on these toxic effects have not been established. This can be explained by the fact that the RYR supplements contain a wide variety of chemical compounds, most of which have not been toxicologically characterized. To fully assess the safety profile of RYR supplements, more research is needed into all these chemical constituents. Based on remaining uncertainties on the effects of RYR supplements, it remains impossible to identify recommended dietary intake levels of monacolins from RYR that would not give any harmful effect to human health. COMPETING INTERESTS There are no competing interests to declare. CONTRIBUTORS All authors substantially contributed to designing the study, acquiring, analyzing and interpreting the data; and drafting and revising the work. All authors approved the submitted final version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. Supporting information TABLE S1 Top 10 reported comedications Click here for additional data file. TABLE S2 Number of reported ADRs per SOC Click here for additional data file. TABLE S3 Overview of all reported cases to Lareb. Click here for additional data file. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
Recovering
ReactionOutcome
CC BY-NC
33085778
19,715,717
2021-04
What was the outcome of reaction 'Pancreatitis'?
Red yeast rice (Monascus purpureus) supplements: Case series assessment of spontaneously reported cases to The Netherlands Pharmacovigilance Centre Lareb. Serious concerns are expressed on the safe use of red yeast rice (RYR) supplements. The aim of the present study was to analyse cases received by Lareb on RYR-related adverse health events. These cases were analysed for the number of reports, number of adverse drug reactions (ADRs), causality, seriousness of the reaction, latency-period, age and sex of the patients, concomitant medication and type of reporter. A total of 94 individual reports were collected by Lareb, corresponding with 187 ADRs. The analysis showed most reported ADRs were labelled musculoskeletal and connective tissue disorders (n = 64), gastrointestinal disorders (n = 33) and general disorders and administration site conditions (n = 23). The use of RYR supplements should be considered as a significant safety concern. Exposure to monacolin K could lead to serious adverse effects. To fully assess the safety profile of RYR supplements, more research is necessary to compose a complete ADR profile of RYR supplements. What is already known about this subject Red yeast rice (RYR) supplements are used to lower serum cholesterol levels in patients with statin intolerance. RYR supplements contain a wide variety of monacolin compounds, of which monacolin K is the primary form being chemically identical to lovastatin. Serious concerns exist on the safety of RYR supplements, for which general toxicological knowledge on the potential health risks and involved mechanism is lacking. What this study adds The present study reveals numerous cases of adverse health effects by RYR and proposes potential toxicological mechanisms involved. Our study confirms that the use of RYR supplements should be considered as a significant safety concern. 1 INTRODUCTION Statins belong to the first‐line therapy in the reduction of elevated blood cholesterol levels and have been shown to reduce the rate of cardiovascular events and reduce all‐time mortality. 1 , 2 However, statins have been associated with muscle‐related adverse events that are most commonly reported as a major reason for statin intolerance 3 Several patients with statin intolerance seek alternative products to reduce elevated blood cholesterol levels. Additionally, even healthy consumers use alternative products to lower serum cholesterol. One of the most common alternative products are red yeast rice (RYR) supplements 3 RYR is produced by fermenting rice with Monascus porpureus, forming monacolin compounds 1 The primary monacolin compound present in the RYR supplements is monacolin K, which is chemically identical to the pharmaceutical lovastatin 4 Monacolin K inhibits HMG‐CoA reductase, an enzyme that catalyses the conversion of HMG‐CoA to mevalonate, a precursor of cholesterol. Through the inhibition of HMG‐CoA, monacolin K decreases the production rate of cholesterol 1 , 5 Several clinical trials have shown that RYR supplements are associated with the prevention of primary and secondary cardiovascular events 4 , 6 , 7 , 8 Besides these effects, also anticancer, anti‐inflammatory, antihypertensive and antidiabetic effects have been described 5 , 9 In contrast to these beneficial health effects, serious concerns exist on the safety of RYR supplements. The structural similarity with lovastatin implies that similar adverse drug reactions (ADRs) can be expected. Overall, general toxicological knowledge on the potential health risks of RYR are lacking. Identification of potential risks of RYR products is challenging because food supplements are not usually registered in pharmacovigilance databases where suspected ADRs can be collected. However, The Netherlands Pharmacovigilance Centre Lareb has received numerous spontaneous reports of ADRs by RYR supplements over recent years. The aim of this article is to comprehensively describe the cases received and assess the case series concerning the use of RYR and health complications. 2 METHODS 2.1 Description of cases In the Netherlands, the Pharmacovigilance Centre Lareb maintains the spontaneous reporting system for drugs, vaccines and natural products with a health claim, such as herbal drugs. By collecting and analysing reported ADRs, knowledge is generated on the occurrence of ADRs in daily practice. Data on reported ADRs was gathered from 1991 (establishment of the centre) until March 2020. The reports can originate from health care professionals or consumers and can be received directly or through pharmaceutical companies. All directly received reports in the Dutch spontaneous reporting system are individually triaged and assessed by trained assessors at Lareb. For the purpose of this study additional causality assessed was performed by the authors according to the World Health Organization causality model to determine the role of RYR in ADR occurrence 10 ADRs are coded with the Medical Dictionary for Regulatory Activities (MedDRA) 11 In the case of unregistered products, Lareb uses a custom built drug dictionary for coding. 12 The number of reports, number of ADRs, seriousness of the reaction 13 latency‐period, age and sex of the patient, concomitant medication, and type of reporter were described. The reports were grouped based on the MedDRA system organ class (SOC) and preferred term (PT). A line‐listing of all selected cases was made. 3 RESULTS 3.1 Characteristics of reports From December 2007 to March 2020, a total of 94 individual reports were collected by Lareb, of which only 16 were received before June 2017. In these reports, in total 187 ADRs were reported, coded as PTs according the standardized MedDRA system. Six reports were according the CIOMS criteria classified as serious: 3 times (acute) pancreatitis and 2 times symptoms of rhabdomyolyses such as chromaturia, abnormal urine odor, myalgia, muscle spasms and malaise were reported. One report concerned acute hepatic failure with jaundice where the liver transplantation was required. World Health Organization causality assessment revealed the role of RYR in to be certain (n = 2), probably/likely (n = 24), possible (n = 61) and unlikely (n = 7) in the reported cases. The majority of reports (n = 75) were submitted by consumers or other non‐health professionals. Nine cases were reported by pharmacists, 8 cases by a physician and 2 reports were reported by both pharmacist and physician. Reported sex was 60 females and 34 males and the mean age was 64 years (range 17–81 years), the median age was 65 years. Concomitant medication use was reported in 55 reports (59.1%). The most reported comedication was vitamin D (colecalciferol), reported 11 times, followed by clopidogrel (Table S1). It must be noted that 1 report can contain multiple concomitant drugs. 3.2 Action and outcome In 66 reports, patients discontinued their treatment with RYR supplements; in 47 cases of these the symptoms resolved, were recovering or recovered with sequel. The following actions with RYR supplement occurred in the remaining patients: no adjustment (n = 13), unknown (n = 5), dose reduced (n = 2), not applicable (n = 6) and not reported (n = 2). 3.3 SOC reports A total of 94 individual reports of ADRs, corresponding to 187 PTs were related to RYR supplement use (Table S3). The most frequently reported PTs were labelled under the SOC musculoskeletal and connective tissue disorders, gastrointestinal disorders, general disorders and administration site conditions and nervous system disorders (Table S2). In total 64 PTs were reported in the SOC musculoskeletal and connective tissue disorders, which included myalgia, muscle spasms, weakness rhabdomyolysis, muscle atrophy, limb discomfort, flank pain, osteoarthritis and tendonitis. Gastrointestinal disorders was reported 33 times, and included inter alia (i.a.) abdominal pain, abdominal discomfort, nausea, diarrhoea and pancreatitis. General disorders and administration site conditions was reported 23 times and included i.a. fatigue, malaise and peripheral oedema. Nervous system disorders, such as headache, dizziness and sleep disorders, were reported 16 times. All SOCs and related no. of ADRs can be found in Table S2. 3.4 Latency Notable the median latency time for all reported ADRs is 2.1 weeks and also median latency's for the symptoms classified within the 4 most reported SOCs show the same tendency, as it falls between 1.9 weeks for nervous system disorders, 2 weeks for general symptoms, 2.4 weeks for musculoskeletal disorders and 2.9 weeks for gastrointestinal disorders (Figure 1B). FIGURE 1 Median latency times for the symptoms in the cases reported to Lareb. GD: general disorders and administration site conditions; NSD: nervous system disorders; GID: gastrointestinal disorders; MCD: musculoskeletal and connective tissue disorders; SOCs: system organ classes 4 DISCUSSION Although RYR‐containing products have been available on the market as food supplements for several years and their efficacy has been proven, 4 , 6 , 7 , 8 serious concerns about their safety profile remain. The present study reveals the most reported ADRs due to the use of RYR supplements to Lareb. It can be noted that the profile of reported ADRs to RYR of the present study is in line with previous studies, 14 , 15 , 16 ] with musculoskeletal and gastrointestinal, as the most frequently reported adverse effects (AEs). Although similar studies have been published before, these supplements are being used on a large scale and new cases of AEs due to these supplements arise. Our case series adds to the existing case reports and contributes to increasing the awareness for the potential adverse health effects of RYR supplements. Overall, our results demonstrate that the safety profile of RYR supplements is similar to that of the synthetic statins, particularly lovastatin. Several uncertainties exist that complicate drawing a complete safety profile of RYR supplements. First of all, there is a wide variability in composition of RYR‐containing food supplements. The monacolins in RYR are used in multi‐ingredient botanical preparations containing multiple other components, which have not been fully evaluated individually or in combination. Moreover, it is evident that the ratio of monacolin K and its hydroxyl acid form monacolin KA differs widely in numerous RYR products. 17 Other than monacolin K, there is a lack of data on the bioactivity of the other components in RYR. Therefore, it is difficult to assess the toxicological mechanisms of this mixture of monacolins. Since RYR most abundantly consists of monacolin K, which is a structural homologue to lovastatin, the mechanisms of RYR‐induced adverse effects are expected to be similar to those known for statins. 4.1 Musculoskeletal disorders Skeletal muscle abnormalities are the most reported and clinically important side effects of RYR, as shown in Table S2. Smith et al. published a case report of a middle‐ aged man presenting with muscle weakness after receiving RYR supplements for 2 months. 18 After examining the patients, they found increased creatine kinase levels of 385 IU/L (normal range is 30–160 IU/L). Additionally, Venhuis et al. reported a similar case report of patients using RYR products. 19 The patients exhibited multiple AEs, including myalgia. Although the exact mechanisms behind the musculoskeletal ADRs are not known, numerous studies have been performed on identification of the mechanism. 20 , 21 Multiple proposed mechanisms include inhibition of coenzyme Q10 synthesis, isoprenoid depletion, decreased or altered sarcolemma membrane cholesterol, disturbed calcium metabolism or autoimmune phenomena 22 , 23 , 24 , 25 The most obvious explanation for RYR‐induced myopathy would be the inhibition of mevalonate, a precursor of coenzyme Q10, due to the HMG‐CoA inhibition. As a consequence of this inhibition, the mitochondrial respiratory chain may be impaired, and this may result in impaired energy production and ultimately cell death 26 This effect is better described for statins, as several studies have shown lower levels of coenzyme Q10 after the use of statins 27 , 28 4.2 Gastrointestinal related disorders The cases described by the present study show that gastrointestinal reactions are often associated with the use of RYR supplements. These symptoms, varying from dyspepsia to vomiting and abdominal pain, nausea and diarrhoea, are also mostly reported by other studies 6 Mazzanti et al. also found a high percentage of gastrointestinal disorders: 22% of all reported ADRs. In general, the reactions were not serious with the exception of 1 hospitalization 15 Although there is no specific explanation yet for these side effects of RYR supplements, gastrointestinal reactions are not uncommon reactions associated with the use of statins, in which mitochondrial dysfunction is suggested to play an important role 29 , 30 4.3 Hepatic disorders Besides musculoskeletal and gastrointestinal disorders, adverse effects associated with RYR that are repeatedly reported are elevated levels of hepatic enzymes, indicative for liver injury. This is also shown by a meta‐analysis on the efficacy and safety of RYR supplements that included a total of 13 randomized controlled trials 31 The analysis showed that the serum alanine transaminase and aspartate aminotransferase levels were significantly increased in RYR users. This might explain the 3 cases of liver injury identified by the present study. In addition to the 3 cases reported by our study, multiple other cases of RYR‐induced liver toxicity have been documented 14 , 15 Three individual cases of severe hepatitis due to RYR were published previously, highlighting that, despite the low incidence of liver injury, the use of RYR supplements might have serious health consequences 32 , 33 , 34 The exact mechanism by which RYR causes hepatotoxicity is not well understood yet. However, it is expected that liver injury by RYR supplements may be attributed to the presence of lovastatin, as liver injuries are also among the most reported side effects of statin users 1 , 35 , 36 , 37 Similar to the musculoskeletal adverse effects, mitochondrial dysfunction is proposed to play a major role in the observed liver injury by statins 38 , 39 4.4 Kidney damage In our case series, kidney damage due to the use of RYR supplements was reported 5 times. The induced kidney damage might be explained by the presence of the mycotoxin citrinin in a significant number of RYR supplement, although this is unfortunately not measured 40 Citrinin is known to have nephrotoxic properties and therefore RYR supplements containing citrinin is a cause for concern, since it can pose a serious health risk: both in vivo and in vitro studies showed that citrinin has potentially mutagenic and mutatoxic properties 41 , 42 , 43 , 44 Another possible explanation for the toxic effects on the kidneys can be found in the muscle damage by RYR supplements. This can result in apoptosis of muscle cells, causing release myoglobin into the blood stream. In the kidneys, myoglobin can be converted to ferrihaem, which is considered a nephrotoxic compound 45 4.5 Possible interactions between comedication and RYR supplements Comedication influencing the bioavailability of monacolin K could also increase the chance of adverse health events by RYR supplements. The present case series shows that approximately 60% of the reported cases concerned the concomitant use of RYR supplements and medicines (Table S1). The use of comedication can potentially interfere with the kinetics and dynamics of RYR. Statins are metabolized by cytochrome P450 3A4 enzymes, and the administration of RYR with cytochrome enzyme inhibitors can therefore increase the risk of side effects by increasing monacolin K plasma concentrations. Such pharmacokinetic interaction has been reported in a 28‐year‐old kidney transplantation female patient who used a preparation containing RYR and cyclosporine 46 This patient developed rhabdomyolysis several months after starting the use of RYR, after receiving a combination of drugs in her post‐transplant period. Another case report suggests that coadministration of RYR with esomeprazole may increase the plasma concentrations of monacolin K and thus the associated risk of myopathy. The proposed mechanism is competitive inhibition of intestinal P‐glycoprotein, resulting in decreased drug secretion into the intestinal lumen and increased drug bioavailability 47 Also, pharmacodynamic interactions may occur by the combined use of drugs and RYR supplements. This will especially be relevant in case of taking RYR together with other statins. In 2 of our cases, atorvastatin was reported as comedication. This means that a potentiation of the HMCG‐CoA inhibition may occur, potentially leading to an increased risk for the occurrence of the ADRs. Hence, the use of RYR in combination with other drugs needs to be done carefully due to potential interactions that may lead to serious adverse health effects. 5 LIMITATIONS In the assessment of case series of spontaneously reported cases, several limitations could play a role. Whereas comedications potentially cause interactions with RYR supplements and thereby increase the chance of developing ADRs, they may also have a confounding effect that complicates identifying the role of RYR in the ADRs. There are, for instance, concomitant drugs known to be potentially associated with pancreatitis, such as morphine, perindopril and telmisartan. To exclude this confounding effect of drugs as much as possible, the use of comedications is taken into account in the causality assessment. Another potential limitation of the present case series of spontaneous reports is notoriety bias 48 Although notoriety bias always plays a role when analysing and discussing spontaneous reports, this does not mean that the reported cases to Lareb are no real cases, only that the reporter was triggered to submit their case after the media attention. Seeing that the cases are reported over a period of multiple years, the increase in reports is likely to reflect the growing use of RYR products and is not merely based on more media attention. Finally, the absence of information about content of monacolin K makes it difficult to strongly relate the ADRs to the use of RYR supplements. However, even if such information were available, this would still provide uncertain information for 2 reasons: (i) based on the reports, it cannot be determined how many dosages 1 patient used; (ii) as the supplements were generally not chemically analysed, it remains uncertain what exactly, and how much, is present of monacolins in the supplement. Interestingly, the analysis of 1 RYR sample of 1 of the reports showed that the dose of monacolin K in the supplements varied between pills from 1 jar (7 vs 3 mg). It is therefore impossible to assess the chemicals and doses to which the patients were exposed. 6 CONCLUSION The intake of monacolins via dietary supplements can lead to serious adverse health effects. Monacolin K is chemically identical to lovastatin and therefore these adverse health effects are similar to those of lovastatin. The use of RYR supplements should therefore be considered as a significant safety concern. So far, exact mechanisms on these toxic effects have not been established. This can be explained by the fact that the RYR supplements contain a wide variety of chemical compounds, most of which have not been toxicologically characterized. To fully assess the safety profile of RYR supplements, more research is needed into all these chemical constituents. Based on remaining uncertainties on the effects of RYR supplements, it remains impossible to identify recommended dietary intake levels of monacolins from RYR that would not give any harmful effect to human health. COMPETING INTERESTS There are no competing interests to declare. CONTRIBUTORS All authors substantially contributed to designing the study, acquiring, analyzing and interpreting the data; and drafting and revising the work. All authors approved the submitted final version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. Supporting information TABLE S1 Top 10 reported comedications Click here for additional data file. TABLE S2 Number of reported ADRs per SOC Click here for additional data file. TABLE S3 Overview of all reported cases to Lareb. Click here for additional data file. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
Recovered
ReactionOutcome
CC BY-NC
33085778
19,715,798
2021-04
What was the outcome of reaction 'Potentiating drug interaction'?
Red yeast rice (Monascus purpureus) supplements: Case series assessment of spontaneously reported cases to The Netherlands Pharmacovigilance Centre Lareb. Serious concerns are expressed on the safe use of red yeast rice (RYR) supplements. The aim of the present study was to analyse cases received by Lareb on RYR-related adverse health events. These cases were analysed for the number of reports, number of adverse drug reactions (ADRs), causality, seriousness of the reaction, latency-period, age and sex of the patients, concomitant medication and type of reporter. A total of 94 individual reports were collected by Lareb, corresponding with 187 ADRs. The analysis showed most reported ADRs were labelled musculoskeletal and connective tissue disorders (n = 64), gastrointestinal disorders (n = 33) and general disorders and administration site conditions (n = 23). The use of RYR supplements should be considered as a significant safety concern. Exposure to monacolin K could lead to serious adverse effects. To fully assess the safety profile of RYR supplements, more research is necessary to compose a complete ADR profile of RYR supplements. What is already known about this subject Red yeast rice (RYR) supplements are used to lower serum cholesterol levels in patients with statin intolerance. RYR supplements contain a wide variety of monacolin compounds, of which monacolin K is the primary form being chemically identical to lovastatin. Serious concerns exist on the safety of RYR supplements, for which general toxicological knowledge on the potential health risks and involved mechanism is lacking. What this study adds The present study reveals numerous cases of adverse health effects by RYR and proposes potential toxicological mechanisms involved. Our study confirms that the use of RYR supplements should be considered as a significant safety concern. 1 INTRODUCTION Statins belong to the first‐line therapy in the reduction of elevated blood cholesterol levels and have been shown to reduce the rate of cardiovascular events and reduce all‐time mortality. 1 , 2 However, statins have been associated with muscle‐related adverse events that are most commonly reported as a major reason for statin intolerance 3 Several patients with statin intolerance seek alternative products to reduce elevated blood cholesterol levels. Additionally, even healthy consumers use alternative products to lower serum cholesterol. One of the most common alternative products are red yeast rice (RYR) supplements 3 RYR is produced by fermenting rice with Monascus porpureus, forming monacolin compounds 1 The primary monacolin compound present in the RYR supplements is monacolin K, which is chemically identical to the pharmaceutical lovastatin 4 Monacolin K inhibits HMG‐CoA reductase, an enzyme that catalyses the conversion of HMG‐CoA to mevalonate, a precursor of cholesterol. Through the inhibition of HMG‐CoA, monacolin K decreases the production rate of cholesterol 1 , 5 Several clinical trials have shown that RYR supplements are associated with the prevention of primary and secondary cardiovascular events 4 , 6 , 7 , 8 Besides these effects, also anticancer, anti‐inflammatory, antihypertensive and antidiabetic effects have been described 5 , 9 In contrast to these beneficial health effects, serious concerns exist on the safety of RYR supplements. The structural similarity with lovastatin implies that similar adverse drug reactions (ADRs) can be expected. Overall, general toxicological knowledge on the potential health risks of RYR are lacking. Identification of potential risks of RYR products is challenging because food supplements are not usually registered in pharmacovigilance databases where suspected ADRs can be collected. However, The Netherlands Pharmacovigilance Centre Lareb has received numerous spontaneous reports of ADRs by RYR supplements over recent years. The aim of this article is to comprehensively describe the cases received and assess the case series concerning the use of RYR and health complications. 2 METHODS 2.1 Description of cases In the Netherlands, the Pharmacovigilance Centre Lareb maintains the spontaneous reporting system for drugs, vaccines and natural products with a health claim, such as herbal drugs. By collecting and analysing reported ADRs, knowledge is generated on the occurrence of ADRs in daily practice. Data on reported ADRs was gathered from 1991 (establishment of the centre) until March 2020. The reports can originate from health care professionals or consumers and can be received directly or through pharmaceutical companies. All directly received reports in the Dutch spontaneous reporting system are individually triaged and assessed by trained assessors at Lareb. For the purpose of this study additional causality assessed was performed by the authors according to the World Health Organization causality model to determine the role of RYR in ADR occurrence 10 ADRs are coded with the Medical Dictionary for Regulatory Activities (MedDRA) 11 In the case of unregistered products, Lareb uses a custom built drug dictionary for coding. 12 The number of reports, number of ADRs, seriousness of the reaction 13 latency‐period, age and sex of the patient, concomitant medication, and type of reporter were described. The reports were grouped based on the MedDRA system organ class (SOC) and preferred term (PT). A line‐listing of all selected cases was made. 3 RESULTS 3.1 Characteristics of reports From December 2007 to March 2020, a total of 94 individual reports were collected by Lareb, of which only 16 were received before June 2017. In these reports, in total 187 ADRs were reported, coded as PTs according the standardized MedDRA system. Six reports were according the CIOMS criteria classified as serious: 3 times (acute) pancreatitis and 2 times symptoms of rhabdomyolyses such as chromaturia, abnormal urine odor, myalgia, muscle spasms and malaise were reported. One report concerned acute hepatic failure with jaundice where the liver transplantation was required. World Health Organization causality assessment revealed the role of RYR in to be certain (n = 2), probably/likely (n = 24), possible (n = 61) and unlikely (n = 7) in the reported cases. The majority of reports (n = 75) were submitted by consumers or other non‐health professionals. Nine cases were reported by pharmacists, 8 cases by a physician and 2 reports were reported by both pharmacist and physician. Reported sex was 60 females and 34 males and the mean age was 64 years (range 17–81 years), the median age was 65 years. Concomitant medication use was reported in 55 reports (59.1%). The most reported comedication was vitamin D (colecalciferol), reported 11 times, followed by clopidogrel (Table S1). It must be noted that 1 report can contain multiple concomitant drugs. 3.2 Action and outcome In 66 reports, patients discontinued their treatment with RYR supplements; in 47 cases of these the symptoms resolved, were recovering or recovered with sequel. The following actions with RYR supplement occurred in the remaining patients: no adjustment (n = 13), unknown (n = 5), dose reduced (n = 2), not applicable (n = 6) and not reported (n = 2). 3.3 SOC reports A total of 94 individual reports of ADRs, corresponding to 187 PTs were related to RYR supplement use (Table S3). The most frequently reported PTs were labelled under the SOC musculoskeletal and connective tissue disorders, gastrointestinal disorders, general disorders and administration site conditions and nervous system disorders (Table S2). In total 64 PTs were reported in the SOC musculoskeletal and connective tissue disorders, which included myalgia, muscle spasms, weakness rhabdomyolysis, muscle atrophy, limb discomfort, flank pain, osteoarthritis and tendonitis. Gastrointestinal disorders was reported 33 times, and included inter alia (i.a.) abdominal pain, abdominal discomfort, nausea, diarrhoea and pancreatitis. General disorders and administration site conditions was reported 23 times and included i.a. fatigue, malaise and peripheral oedema. Nervous system disorders, such as headache, dizziness and sleep disorders, were reported 16 times. All SOCs and related no. of ADRs can be found in Table S2. 3.4 Latency Notable the median latency time for all reported ADRs is 2.1 weeks and also median latency's for the symptoms classified within the 4 most reported SOCs show the same tendency, as it falls between 1.9 weeks for nervous system disorders, 2 weeks for general symptoms, 2.4 weeks for musculoskeletal disorders and 2.9 weeks for gastrointestinal disorders (Figure 1B). FIGURE 1 Median latency times for the symptoms in the cases reported to Lareb. GD: general disorders and administration site conditions; NSD: nervous system disorders; GID: gastrointestinal disorders; MCD: musculoskeletal and connective tissue disorders; SOCs: system organ classes 4 DISCUSSION Although RYR‐containing products have been available on the market as food supplements for several years and their efficacy has been proven, 4 , 6 , 7 , 8 serious concerns about their safety profile remain. The present study reveals the most reported ADRs due to the use of RYR supplements to Lareb. It can be noted that the profile of reported ADRs to RYR of the present study is in line with previous studies, 14 , 15 , 16 ] with musculoskeletal and gastrointestinal, as the most frequently reported adverse effects (AEs). Although similar studies have been published before, these supplements are being used on a large scale and new cases of AEs due to these supplements arise. Our case series adds to the existing case reports and contributes to increasing the awareness for the potential adverse health effects of RYR supplements. Overall, our results demonstrate that the safety profile of RYR supplements is similar to that of the synthetic statins, particularly lovastatin. Several uncertainties exist that complicate drawing a complete safety profile of RYR supplements. First of all, there is a wide variability in composition of RYR‐containing food supplements. The monacolins in RYR are used in multi‐ingredient botanical preparations containing multiple other components, which have not been fully evaluated individually or in combination. Moreover, it is evident that the ratio of monacolin K and its hydroxyl acid form monacolin KA differs widely in numerous RYR products. 17 Other than monacolin K, there is a lack of data on the bioactivity of the other components in RYR. Therefore, it is difficult to assess the toxicological mechanisms of this mixture of monacolins. Since RYR most abundantly consists of monacolin K, which is a structural homologue to lovastatin, the mechanisms of RYR‐induced adverse effects are expected to be similar to those known for statins. 4.1 Musculoskeletal disorders Skeletal muscle abnormalities are the most reported and clinically important side effects of RYR, as shown in Table S2. Smith et al. published a case report of a middle‐ aged man presenting with muscle weakness after receiving RYR supplements for 2 months. 18 After examining the patients, they found increased creatine kinase levels of 385 IU/L (normal range is 30–160 IU/L). Additionally, Venhuis et al. reported a similar case report of patients using RYR products. 19 The patients exhibited multiple AEs, including myalgia. Although the exact mechanisms behind the musculoskeletal ADRs are not known, numerous studies have been performed on identification of the mechanism. 20 , 21 Multiple proposed mechanisms include inhibition of coenzyme Q10 synthesis, isoprenoid depletion, decreased or altered sarcolemma membrane cholesterol, disturbed calcium metabolism or autoimmune phenomena 22 , 23 , 24 , 25 The most obvious explanation for RYR‐induced myopathy would be the inhibition of mevalonate, a precursor of coenzyme Q10, due to the HMG‐CoA inhibition. As a consequence of this inhibition, the mitochondrial respiratory chain may be impaired, and this may result in impaired energy production and ultimately cell death 26 This effect is better described for statins, as several studies have shown lower levels of coenzyme Q10 after the use of statins 27 , 28 4.2 Gastrointestinal related disorders The cases described by the present study show that gastrointestinal reactions are often associated with the use of RYR supplements. These symptoms, varying from dyspepsia to vomiting and abdominal pain, nausea and diarrhoea, are also mostly reported by other studies 6 Mazzanti et al. also found a high percentage of gastrointestinal disorders: 22% of all reported ADRs. In general, the reactions were not serious with the exception of 1 hospitalization 15 Although there is no specific explanation yet for these side effects of RYR supplements, gastrointestinal reactions are not uncommon reactions associated with the use of statins, in which mitochondrial dysfunction is suggested to play an important role 29 , 30 4.3 Hepatic disorders Besides musculoskeletal and gastrointestinal disorders, adverse effects associated with RYR that are repeatedly reported are elevated levels of hepatic enzymes, indicative for liver injury. This is also shown by a meta‐analysis on the efficacy and safety of RYR supplements that included a total of 13 randomized controlled trials 31 The analysis showed that the serum alanine transaminase and aspartate aminotransferase levels were significantly increased in RYR users. This might explain the 3 cases of liver injury identified by the present study. In addition to the 3 cases reported by our study, multiple other cases of RYR‐induced liver toxicity have been documented 14 , 15 Three individual cases of severe hepatitis due to RYR were published previously, highlighting that, despite the low incidence of liver injury, the use of RYR supplements might have serious health consequences 32 , 33 , 34 The exact mechanism by which RYR causes hepatotoxicity is not well understood yet. However, it is expected that liver injury by RYR supplements may be attributed to the presence of lovastatin, as liver injuries are also among the most reported side effects of statin users 1 , 35 , 36 , 37 Similar to the musculoskeletal adverse effects, mitochondrial dysfunction is proposed to play a major role in the observed liver injury by statins 38 , 39 4.4 Kidney damage In our case series, kidney damage due to the use of RYR supplements was reported 5 times. The induced kidney damage might be explained by the presence of the mycotoxin citrinin in a significant number of RYR supplement, although this is unfortunately not measured 40 Citrinin is known to have nephrotoxic properties and therefore RYR supplements containing citrinin is a cause for concern, since it can pose a serious health risk: both in vivo and in vitro studies showed that citrinin has potentially mutagenic and mutatoxic properties 41 , 42 , 43 , 44 Another possible explanation for the toxic effects on the kidneys can be found in the muscle damage by RYR supplements. This can result in apoptosis of muscle cells, causing release myoglobin into the blood stream. In the kidneys, myoglobin can be converted to ferrihaem, which is considered a nephrotoxic compound 45 4.5 Possible interactions between comedication and RYR supplements Comedication influencing the bioavailability of monacolin K could also increase the chance of adverse health events by RYR supplements. The present case series shows that approximately 60% of the reported cases concerned the concomitant use of RYR supplements and medicines (Table S1). The use of comedication can potentially interfere with the kinetics and dynamics of RYR. Statins are metabolized by cytochrome P450 3A4 enzymes, and the administration of RYR with cytochrome enzyme inhibitors can therefore increase the risk of side effects by increasing monacolin K plasma concentrations. Such pharmacokinetic interaction has been reported in a 28‐year‐old kidney transplantation female patient who used a preparation containing RYR and cyclosporine 46 This patient developed rhabdomyolysis several months after starting the use of RYR, after receiving a combination of drugs in her post‐transplant period. Another case report suggests that coadministration of RYR with esomeprazole may increase the plasma concentrations of monacolin K and thus the associated risk of myopathy. The proposed mechanism is competitive inhibition of intestinal P‐glycoprotein, resulting in decreased drug secretion into the intestinal lumen and increased drug bioavailability 47 Also, pharmacodynamic interactions may occur by the combined use of drugs and RYR supplements. This will especially be relevant in case of taking RYR together with other statins. In 2 of our cases, atorvastatin was reported as comedication. This means that a potentiation of the HMCG‐CoA inhibition may occur, potentially leading to an increased risk for the occurrence of the ADRs. Hence, the use of RYR in combination with other drugs needs to be done carefully due to potential interactions that may lead to serious adverse health effects. 5 LIMITATIONS In the assessment of case series of spontaneously reported cases, several limitations could play a role. Whereas comedications potentially cause interactions with RYR supplements and thereby increase the chance of developing ADRs, they may also have a confounding effect that complicates identifying the role of RYR in the ADRs. There are, for instance, concomitant drugs known to be potentially associated with pancreatitis, such as morphine, perindopril and telmisartan. To exclude this confounding effect of drugs as much as possible, the use of comedications is taken into account in the causality assessment. Another potential limitation of the present case series of spontaneous reports is notoriety bias 48 Although notoriety bias always plays a role when analysing and discussing spontaneous reports, this does not mean that the reported cases to Lareb are no real cases, only that the reporter was triggered to submit their case after the media attention. Seeing that the cases are reported over a period of multiple years, the increase in reports is likely to reflect the growing use of RYR products and is not merely based on more media attention. Finally, the absence of information about content of monacolin K makes it difficult to strongly relate the ADRs to the use of RYR supplements. However, even if such information were available, this would still provide uncertain information for 2 reasons: (i) based on the reports, it cannot be determined how many dosages 1 patient used; (ii) as the supplements were generally not chemically analysed, it remains uncertain what exactly, and how much, is present of monacolins in the supplement. Interestingly, the analysis of 1 RYR sample of 1 of the reports showed that the dose of monacolin K in the supplements varied between pills from 1 jar (7 vs 3 mg). It is therefore impossible to assess the chemicals and doses to which the patients were exposed. 6 CONCLUSION The intake of monacolins via dietary supplements can lead to serious adverse health effects. Monacolin K is chemically identical to lovastatin and therefore these adverse health effects are similar to those of lovastatin. The use of RYR supplements should therefore be considered as a significant safety concern. So far, exact mechanisms on these toxic effects have not been established. This can be explained by the fact that the RYR supplements contain a wide variety of chemical compounds, most of which have not been toxicologically characterized. To fully assess the safety profile of RYR supplements, more research is needed into all these chemical constituents. Based on remaining uncertainties on the effects of RYR supplements, it remains impossible to identify recommended dietary intake levels of monacolins from RYR that would not give any harmful effect to human health. COMPETING INTERESTS There are no competing interests to declare. CONTRIBUTORS All authors substantially contributed to designing the study, acquiring, analyzing and interpreting the data; and drafting and revising the work. All authors approved the submitted final version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. Supporting information TABLE S1 Top 10 reported comedications Click here for additional data file. TABLE S2 Number of reported ADRs per SOC Click here for additional data file. TABLE S3 Overview of all reported cases to Lareb. Click here for additional data file. DATA AVAILABILITY STATEMENT The data that support the findings of this study are available from the corresponding author upon reasonable request.
Recovering
ReactionOutcome
CC BY-NC
33085778
19,715,717
2021-04
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Aphasia'.
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
CISPLATIN, ETOPOSIDE
DrugsGivenReaction
CC BY-NC-ND
33087671
18,566,076
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Carotid artery occlusion'.
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
CISPLATIN, ETOPOSIDE
DrugsGivenReaction
CC BY-NC-ND
33087671
18,566,076
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Cerebral artery occlusion'.
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
CISPLATIN, ETOPOSIDE
DrugsGivenReaction
CC BY-NC-ND
33087671
18,566,076
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Cerebral infarction'.
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
CISPLATIN, ETOPOSIDE
DrugsGivenReaction
CC BY-NC-ND
33087671
18,566,076
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Haemorrhagic cerebral infarction'.
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
CISPLATIN, ETOPOSIDE
DrugsGivenReaction
CC BY-NC-ND
33087671
18,476,765
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Haemorrhagic infarction'.
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
CISPLATIN, ETOPOSIDE
DrugsGivenReaction
CC BY-NC-ND
33087671
18,566,076
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Hemiparesis'.
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
CISPLATIN, ETOPOSIDE
DrugsGivenReaction
CC BY-NC-ND
33087671
18,566,076
2021-03-15
What is the weight of the patient?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
67.9 kg.
Weight
CC BY-NC-ND
33087671
18,476,765
2021-03-15
What was the administration route of drug 'CISPLATIN'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY-NC-ND
33087671
18,476,765
2021-03-15
What was the dosage of drug 'RADIATION THERAPY'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
60 GY
DrugDosageText
CC BY-NC-ND
33087671
19,243,011
2021-03-15
What was the outcome of reaction 'Aortic thrombosis'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Recovered
ReactionOutcome
CC BY-NC-ND
33087671
18,566,076
2021-03-15
What was the outcome of reaction 'Carotid artery occlusion'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Not recovered
ReactionOutcome
CC BY-NC-ND
33087671
18,566,076
2021-03-15
What was the outcome of reaction 'Cerebral artery occlusion'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Not recovered
ReactionOutcome
CC BY-NC-ND
33087671
18,566,076
2021-03-15
What was the outcome of reaction 'Cerebral infarction'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Not recovered
ReactionOutcome
CC BY-NC-ND
33087671
18,566,076
2021-03-15
What was the outcome of reaction 'Embolic stroke'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Not recovered
ReactionOutcome
CC BY-NC-ND
33087671
19,243,011
2021-03-15
What was the outcome of reaction 'Haemorrhagic cerebral infarction'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Recovered
ReactionOutcome
CC BY-NC-ND
33087671
18,476,765
2021-03-15
What was the outcome of reaction 'Haemorrhagic infarction'?
Embolic Stroke Due to a Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. A 59-year-old woman with small-cell lung carcinoma achieved tumor disappearance after cisplatin-based chemotherapy (CBC) and radiation treatment but subsequently experienced right hemiparesis and aphasia. Brain magnetic resonance imaging revealed a left middle cerebral artery territory acute infarction and left internal carotid artery occlusion. Ultrasonography revealed a mobile thrombus in the left common and internal carotid arteries, and contrast computed tomography revealed a mural thrombus in the ascending aorta. Based on these findings, embolic stroke due to aortic mural thrombus following CBC was diagnosed. Aortic mural thrombus is a rare complication of CBC but carries a risk of embolic stroke. Introduction Small-cell lung carcinoma (SCLC) is generally thought to be the most malignant subtype of lung cancer. The standard treatment is cisplatin-based chemotherapy (CBC) combined with radiation therapy for the limited stage and CBC alone for the extensive stage (1). However, cisplatin use carries a potential risk of thromboembolism (2, 3). Figure 1. Computed tomography (CT) of the chest before and after cisplatin-based chemotherapy. Contrast chest CT prior to chemotherapy (A) shows the lung cancer in the right middle lobe (white arrowhead). Non-contrast chest CT immediately after the final chemoradiotherapy course (B) shows that the tumor in the right middle lobe has completely vanished. R indicates right side A through B. We herein report a patient with SCLC who was successfully treated with CBC and radiation but subsequently experienced an ischemic stroke due to a mural thrombus in the ascending aorta. Aortic mural thrombus, especially in the ascending aorta, is a rare complication of CBC but poses a risk of serious embolic stroke. Case Report A 59-year-old right-handed woman with stage IIIB (T3N2M0) SCLC in the right middle lobe (Fig. 1A) was successfully treated with 4 standard courses of cisplatin-etoposide therapy (cisplatin 80 mg/m2/day on day 1 and etoposide 100 mg/m2/day on days 1-3 every 3 weeks for 4 cycles) combined with a total of 60 Gy of radiation, which achieved complete disappearance of the tumor (Fig. 1B). However, right hemiparesis and aphasia developed two days after the final chemotherapy course. She was admitted to a local hospital but was transferred the next day to our hospital, where she had received her chemotherapy. A neurological examination revealed left conjugate deviation, right complete hemiparesis, including the face, and motor-dominant aphasia; her National Institute of Health stroke scale (NIHSS) score was 20. Tendon reflexes in the right upper and lower extremities were slightly increased, and Babinski reflex was positive on the right side. Vital signs were normal; blood pressure was 136/42 mmHg, heart rate was regular and 72/min, respiratory rate was 14/min, and body temperature was 37.1℃. Brain magnetic resonance imaging (MRI) performed in the local hospital (day 1) showed an acute infarction in the left middle cerebral artery (MCA) territory (Fig. 2A-F), non-terminal occlusion of the left internal carotid artery (ICA) and probable main trunk occlusion of the left MCA (Fig. 2G, H). An imaging mismatch between diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) was evident at the time. Previous contrast-enhanced brain MRI examined before the CBC revealed that the left ICA and left MCA appeared to be normal (Fig. 3). This suggested that the tandem ICA-MCA occlusions might be embolic. Figure 2. Magnetic resonance imaging of the brain on day 1. Note the acute brain infarction in the left middle cerebral artery (MCA) territory, showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic area does not show obvious signal changes on fluid-attenuated inversion recovery, except for the left insula cortex (E, F). Magnetic resonance angiography indicates non-terminal occlusion of the left internal carotid artery and probable main trunk occlusion of the left MCA (G, H). R indicates right side A through H. Figure 3. Contrast-enhanced magnetic resonance imaging of the brain before chemotherapy. Contrast-enhanced T1-weighted coronal (A) and axial (B) images show that the left internal carotid (black arrow) and middle cerebral (white arrow) arteries appear to be normal. R indicates right side A through B. Brain MRI on admission (day 2) showed an acute infarction in the anterior territory of the left MCA (Fig. 4A-F) and complete occlusion of the left ICA-MCA (Fig. 4G, H). The DWI-FLAIR mismatch was not observed anymore. A blood cell count on admission showed moderate anemia and the following findings: white blood cells (WBCs) 3,900/μL, red blood cells (RBCs) 282×104/μL, hemoglobin 8.6 g/dL and platelets 15.8×104/μL. Blood biochemistry revealed slightly elevated levels of glucose (141 mg/dL), HbA1c (7.6%) and triglyceride (206 mg/dL), low levels of high-density lipoprotein cholesterol (30 mg/dL), almost normal levels of low-density lipoprotein cholesterol (122 mg/dL) and normal levels of N-terminal pro-brain natriuretic peptide (72 pg/mL). D-dimer levels were slightly increased (2.4 μg/mL), but protein C levels were normal: protein C antigen 100% (normal range: 70-150%) and protein C activity 114% (normal range: 64-146%). Protein S levels were also normal: protein S antigen 98% (normal range: 65-135%), protein S free antigen 104% (normal range: 60-104%) and protein S activity 99% (normal range: 56-126%). Antithrombin-III levels were unremarkable: 93.5% (normal range: 70-130%). Antiphospholipid antibodies were negative. Physiological function tests were performed on days 3-4. A Holter electrocardiogram showed no atrial fibrillation. Transthoracic echocardiography indicated neither valvular abnormalities nor left atrial enlargement (left atrial diameter: 24 mm), and an additional microbubble test with abdominal compression in substitution for the Valsalva maneuver revealed no right-left shunt. Transesophageal echocardiography was not performed in order to avoid any risk of aspiration pneumonia because of her post-chemotherapy condition. Venous ultrasonography revealed asymptomatic distal deep vein thrombosis in the right fibular vein. Carotid ultrasonography showed a mobile thrombus extending from the left common carotid artery to the ICA. Contrast computed tomography (CT) on day 4 revealed a massive thrombus within the left common carotid and internal carotid arteries (Fig. 5A) and a mural thrombus attached to the calcified lesion of the ascending aorta (Fig. 5B, C), which had not been observed before chemotherapy (Fig. 5D). Brain CT on day 8 demonstrated hemorrhagic infarction in the left MCA territory (Fig. 5E). Based on these findings, cardiogenic embolism, including paradoxical embolism, was unlikely, and aortic mural thrombus was considered a potential embolic source in the patient. Embolic stroke due to an aortic mural thrombus following CBC was therefore diagnosed. Figure 4. Magnetic resonance imaging of the brain on day 2. Note the acute brain infarction in the anterior territory of the left middle cerebral artery (MCA), showing a high signal intensity on diffusion-weighted imaging (A, B) and low signal intensity on apparent diffusion coefficient maps (C, D). The ischemic lesion also exhibits a high signal intensity on fluid-attenuated inversion recovery (E, F). Magnetic resonance angiography indicates complete occlusion of the left internal carotid artery and the left MCA, but cross flow through anterior communicating artery supplies the left anterior cerebral artery (G, H). R indicates right side A through H. Figure 5. Computed tomography (CT) of the brain, neck and chest. Contrast neck CT on day 4 (A) shows the massive thrombus in the left internal carotid artery (white arrow). Contrast chest CT on day 4 (B, C) demonstrates the aortic mural thrombus (black arrow) attached to the calcified lesion of the ascending aorta (white arrowhead). There is no mural thrombus in the ascending aorta on contrast chest CT examined before chemotherapy (D). Brain CT on day 8 (E) displays hemorrhagic infarction in the left middle cerebral artery territory. Contrast chest CT on day 17 (F) shows the disappearance of the aortic mural thrombus. R indicates right side A through F. After admission she received 60 mg/day of intravenous edaravone, a free radical scavenger, and intravenous heparin was begun in order to achieve 1.5-fold prolongation of the activated partial thromboplastin time over the baseline for the aortic mural thrombus. Due to progressive pancytopenia on day 4 (WBCs 1,600/μL, RBCs 253×104/μL, hemoglobin 7.6 g/dL, platelets 7.9×104/μL) resulting from the final course of chemotherapy, surgical thrombectomy was not performed. Contrast CT on day 17 revealed complete disappearance of the aortic mural thrombus (Fig. 5F) with no additional whole-body embolisms observed, and the antithrombotic therapy was changed to aspirin 81 mg/day. She was transferred to a rehabilitation facility on day 42. After six months of rehabilitation, she still exhibited right hemiparesis and motor-dominant aphasia (NIHSS score 13 and modified Rankin scale 4). Brain MRI at 12 months after the stroke onset showed an old infarction in the left MCA territory (Fig. 6A, B), which was basically similar to the previously observed lesion, and occlusion of the left ICA-MCA (Fig. 6C, D). She showed no cancer recurrence or further thromboembolic events for at least 18 months and had a normal D-dimer level despite no anticoagulant use. Figure 6. Magnetic resonance imaging at 12 months after stroke. T2-weighted images (A, B) show old infarction in the left middle cerebral artery (MCA) territory. Magnetic resonance angiography shows occlusions of the left internal carotid artery and the left MCA (C, D). R indicates right side A through D. Discussion Machleder et al. reviewed 10,671 consecutive autopsies and identified 48 cases of nonaneurysmal aortic mural thrombus, of which 38 were in the abdominal aorta, 1 was in the thoracic aorta, and 9 were in both (4). Pagni et al. analyzed 14 patients with symptomatic thoracic aortic mural thrombus and found that only 1 patient had a mural thrombus in the ascending aorta (5). These findings point to the rarity of a nonaneurysmal mural thrombus in the ascending aorta. An ischemic stroke, particularly an embolic stroke, in patients with active cancer may be a sign of Trousseau syndrome, which is thought to arise from the hypercoagulability associated with cancer (6). This disorder generally has a poor prognosis, as seen in the median survival time of 4.5 months (7). Although the present patient suffered from cancer and eventually experienced an ischemic stroke, Trousseau syndrome was unlikely to be the cause of the stroke for the following reasons: first, chemoradiotherapy had achieved complete resolution of the lung tumor prior to the stroke onset; second, no thromboembolic events had occurred for more than 18 months during aspirin therapy following the disappearance of the aortic mural thrombus although the initial treatment began with heparin; finally, the D-dimer level had remained normal despite the discontinuation of anticoagulants. Standard chemotherapy for SCLC consists of a cisplatin-based regimen (1), but cisplatin is known to be a risk factor of thromboembolism. Lee et al. analyzed 277 patients with SCLC who received chemotherapy, of whom 218 received cisplatin, and found that CBC was an independent risk factor of thromboembolism, as indicated by a hazard ratio of 4.36 (2). Moore et al. also analyzed 932 cancer patients treated with CBC and discovered an extremely high incidence of 18.1% for thromboembolisms, most of which were deep vein thromboses and pulmonary embolisms; arterial embolisms were rare (3). Although the pathogenesis of cisplatin-related arterial embolisms remains uncertain, endothelial cell damage, as indicated by von Willebrand factor release, may be a contributing factor (8). Endothelial damage was not confirmed in the present patient, because the von Willebrand factor level was not examined. However, the aortic calcified lesion might suggest atherosclerotic endothelial impairment, and CBC together with atherosclerosis might have generated mural thrombus in the present patient. Thus far, only two cases of aortic mural thrombus associated with CBC in the ascending aorta have been reported (9, 10), and the characteristics of the patients are summarized in Table. A patient reported by Moorjani et al. was undergoing CBC for bladder carcinoma and was incidentally found to have an asymptomatic aortic mural thrombus on three-dimensional CT of the chest (9). Surgical thrombectomy disclosed mobile thrombus adherent to the ascending aorta along with a separate aortic ulcer (9). Similar to our patient, the atherosclerotic endothelial impairment together with cisplatin-induced endothelial damage may have caused the aortic mural thrombus in that patient. Another patient reported by Yagyu et al. also had an asymptomatic aortic mural thrombus after pre-operative CBC for gastric cancer, which was incidentally found on enhanced CT for an evaluation of the response to chemotherapy. That patient also had protein C deficiency, a risk factor of hypercoagulability (10). Both patients were asymptomatic, and neither had an ischemic stroke. Atherosclerosis risk factors were not mentioned in either case. The present patient did not have any coagulation disorders as far as we found but was suggested to have potential atherosclerotic endothelial damage of the ascending aorta. Although arterial thrombosis is a rare complication after CBC, cisplatin-induced endothelial damage in combination with additional factors, such as coagulation disorder and atherosclerosis, may cause aortic mural thrombus. Table. Clinical Characteristics of Patients with Mural Thrombus in the Ascending Aorta Following Cisplatin-based Chemotherapy. Age Sex Cancer Symptoms Coagulation disorder Atherosclerosis Initial treatment Long-term treatment Case 1 (9) 53 yo M BC None Not described Aortic ulcer Surgical thrombectomy Warfarin Case 2 (10) 70 yo M GC None Protein C deficiency Not described Heparin/Warfarin None Present case 59 yo F SCLC IS None Aortic calcification Heparin Aspirin yo: years old, M: male, F: female, BC: bladder carcinoma, GC: gastric carcinoma, SCLC: small cell lung carcinoma, IS: ischemic stroke The present report is the first to describe an embolic stroke due to a mural thrombus attached to the ascending aorta following CBC. Physicians should be aware of aortic mural thrombus as a rare cause of ischemic stroke in patients treated with CBC, given the wide use of cisplatin against various cancers aside from SCLC. The authors state that they have no Conflict of Interest (COI). Acknowledgement The authors are grateful to Mr. James R. Valera for his assistance in editing the manuscript.
Not recovered
ReactionOutcome
CC BY-NC-ND
33087671
18,566,076
2021-03-15
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Anaemia'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
CHOLECALCIFEROL, DARBEPOETIN ALFA, ENALAPRIL, ENOXAPARIN SODIUM, ESOMEPRAZOLE MAGNESIUM, FUROSEMIDE, LEVOTHYROXINE, PENICILLIN V, RANITIDINE, SODIUM CHLORIDE, WARFARIN
DrugsGivenReaction
CC BY
33089377
19,416,137
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Anticoagulation drug level above therapeutic'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ALBUMIN HUMAN, INDOMETHACIN, OMEPRAZOLE SODIUM, WARFARIN
DrugsGivenReaction
CC BY
33089377
18,591,197
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Catheter site haemorrhage'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ALBUMIN HUMAN, INDOMETHACIN, OMEPRAZOLE SODIUM, WARFARIN
DrugsGivenReaction
CC BY
33089377
18,591,197
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Drug interaction'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
CHOLECALCIFEROL, DARBEPOETIN ALFA, ENALAPRIL, ENOXAPARIN SODIUM, ESOMEPRAZOLE MAGNESIUM, FUROSEMIDE, LEVOTHYROXINE, PENICILLIN V, RANITIDINE, SODIUM CHLORIDE, WARFARIN
DrugsGivenReaction
CC BY
33089377
19,416,137
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Haematemesis'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ALBUMIN HUMAN, INDOMETHACIN, OMEPRAZOLE SODIUM, WARFARIN
DrugsGivenReaction
CC BY
33089377
18,591,197
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Haematochezia'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ALBUMIN HUMAN, INDOMETHACIN, OMEPRAZOLE SODIUM, WARFARIN
DrugsGivenReaction
CC BY
33089377
18,591,197
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Haemoglobin decreased'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ALBUMIN HUMAN, INDOMETHACIN, OMEPRAZOLE SODIUM, WARFARIN
DrugsGivenReaction
CC BY
33089377
18,591,197
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'International normalised ratio increased'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ALBUMIN HUMAN, INDOMETHACIN, OMEPRAZOLE SODIUM, WARFARIN
DrugsGivenReaction
CC BY
33089377
18,591,197
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Post procedural haemorrhage'.
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ALBUMIN HUMAN, INDOMETHACIN, OMEPRAZOLE SODIUM, WARFARIN
DrugsGivenReaction
CC BY
33089377
18,591,197
2021-05
What was the administration route of drug 'ALBUMIN HUMAN'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33089377
18,591,197
2021-05
What was the administration route of drug 'DARBEPOETIN ALFA'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33089377
19,416,137
2021-05
What was the administration route of drug 'ENOXAPARIN SODIUM'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Subcutaneous
DrugAdministrationRoute
CC BY
33089377
19,416,137
2021-05
What was the administration route of drug 'FUROSEMIDE'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33089377
19,416,137
2021-05
What was the administration route of drug 'OMEPRAZOLE SODIUM'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33089377
18,591,197
2021-05
What was the dosage of drug 'ESOMEPRAZOLE MAGNESIUM'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
STARTED AFTER FIRST BLEEDING EVENT
DrugDosageText
CC BY
33089377
19,416,137
2021-05
What was the dosage of drug 'SODIUM CHLORIDE'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
1MMOL/ML
DrugDosageText
CC BY
33089377
19,416,137
2021-05
What was the outcome of reaction 'Anaemia'?
Thromboprophylaxis in congenital nephrotic syndrome: 15-year experience from a national cohort. Congenital nephrotic syndrome (CNS) is an ultra-rare disease associated with a pro-thrombotic state and venous thromboembolisms (VTE). There is very limited evidence evaluating thromboprophylaxis in patients with CNS. This study aimed to determine the doses and duration of treatment required to achieve adequate thromboprophylaxis in patients with CNS. From 2005 to 2018 children in Scotland with a confirmed genetic or histological diagnosis of CNS were included if commenced on thromboprophylaxis. The primary study endpoint was stable drug monitoring. Secondary outcomes included VTE or significant haemorrhage. Eight patients were included; all initially were commenced on low-molecular weight heparin (enoxaparin). Four patients maintained therapeutic anti-Factor Xa levels (time 3-26 weeks, dose 3.2-5.07 mg/kg/day), and one patient developed a thrombosis (Anti-Factor Xa: 0.27 IU/ml). Four patients were subsequently treated with warfarin. Two patients maintained therapeutic INRs (time 6-11 weeks, dose 0.22-0.25 mg/kg/day), and one patient had two bleeding events (Bleed 1: INR 6, Bleed 2: INR 5.5). Achieving thromboprophylaxis in CNS is challenging. Similar numbers of patients achieved stable anticoagulation on warfarin and enoxaparin. Enoxaparin dosing was nearly double the recommended starting doses for secondary thromboprophylaxis. Bleeding events were all associated with supra-therapeutic anticoagulation. Introduction Congenital nephrotic syndrome (CNS) is a rare disease characterised by heavy proteinuria and severe oedema developing within 3 months of birth [1, 2]. Glomerular filtration barrier proteins are defective due to genetic mutations or more rarely secondary to congenital viral infection. Complications arising from severe proteinuria include venous thromboembolism (VTE), recurrent infection, fluid and electrolyte disturbance, and impaired growth [3]. The increased VTE risk is predominantly attributed to urinary loss of proteins important in coagulation regulation, exacerbated by the common requirement in this patient group for long-term central venous access [4–6]. Loss of haemostatic proteins, e.g., antithrombin III, leads to an up-regulation in hepatic coagulation factor synthesis and thus a pro-thrombotic tendency [7–10]. Several studies report a VTE prevalence of 10–29% of CNS patients over their disease course; this variability being partly attributed to the marked genotypic and phenotypic variation in CNS [1, 11, 12]. To mitigate the thrombotic risk, management includes strategies to reduce urinary protein loss and administration of anticoagulant therapies. Protein loss is minimised by bilateral nephrectomy and early use of dialysis, or unilateral nephrectomy in combination with angiotensin converting enzyme inhibitors and prostaglandin inhibitors to decrease GFR [4, 13]. Anticoagulation agents commonly used are warfarin and enoxaparin. Warfarin, a vitamin K antagonist, is monitored using the international normalised ratio (INR). The target INR is between 2.0 and 3.0 for primary thromboprophylaxis [14]. Enoxaparin, a low molecular weight heparin (LMWH), binds to anti-thrombin leading to inhibition of activated factor X. Anti-factor Xa assays are used to monitor efficacy, with a target level between 0.2 and 0.4 IU/ml for primary thromboprophylaxis [14, 15]. If a thrombotic event has already occurred, levels are targeted at 0.5–1 IU/ml for secondary thromboprophylaxis. Aspirin is less frequently used as thromboprophylaxis in CNS and is not utilised within our unit. Unfractionated heparin is not suitable as it requires continuous infusion, as well as an extensive adverse effect profile [2]. Direct oral anticoagulants have not been studied in CNS. Thromboprophylaxis in children is challenging due to rapid growth velocity and physiological changes in pharmacokinetics, especially in the early years of life [16, 17]. Fung et al. demonstrated that therapeutic anti-factor Xa levels required an average of 1.64 mg/kg and 1.45 mg/kg of enoxaparin for children under 1 year and aged 1 to 6 years, respectively [16, 18]. Thromboprophylaxis using LMWH in CNS is further complicated by antithrombin III deficiency (due to urinary loss) causing heparin resistance [19]. Warfarin also has challenges in infancy, as metabolism is influenced by comorbidities, medications, and dietary changes. Similar to enoxaparin, higher doses are typically required in infants than children with doses of ~ 0.32 mg/kg and ~ 0.09 mg/kg reported in children under 1 and over 11, respectively [20]. Infants also typically require longer treatments to achieve target INRs and more frequent dose adjustments when compared with older children [21]. The extreme rarity of CNS is a significant limitation on the ability to undertake a clinical trial of thromboprophylaxis. Therapeutic decisions are based on patient preference and clinician experience. In a recent European multi-centre retrospective review of anticoagulation in CNS, 5/45 (11%) patients receiving anticoagulant therapy and 4/26 (15%) not receiving anticoagulants developed VTE (p = 0.60) [22]. Anticoagulant therapies in patients experiencing VTE were warfarin (n = 3), heparin (n = 1), and aspirin (n = 1). Despite participation by 17 tertiary centres, the rarity of CNS and VTE as an outcome precluded formal statistical analysis due to small numbers. Additionally, therapeutic monitoring was not reported, making it uncertain whether VTE occurred due to inadequate thromboprophylaxis in the ‘anticoagulated’ cohort. Our own observation was that patients often required high doses of anticoagulant agents to achieve sufficient therapeutic levels. This case series aims to report whether significantly higher doses of anticoagulants are required to achieve adequate thromboprophylaxis in patients with CNS. We hypothesised that patients will require high doses of anticoagulants with a prolonged time taken to reach therapeutic levels. Methods Data were obtained from patients admitted to the Royal Hospital for Children, Glasgow. Patients were included if CNS was diagnosed from 1 July 2005 until 1 January 2018. The database was locked on 1 June 2020. As a single national paediatric nephrology centre, this represents all CNS cases in Scotland in that time period. The data were collected retrospectively using clinical portal (TrakCare, InterSystems corporation) and the Strathclyde electronic renal patient record (SERPR) (VitalDataClient, v1.6.0.9493). Graphs were produced using GraphPad Prism version 8 (GraphPad Software, San Diego, CA). Data collected included basic demographic data, length, weight, serum creatinine, serum albumin, urinary protein:creatinine ratio, factor Xa assays, INR, antithrombin III levels, thromboprophylaxis dose in mg/kg/day, concomitant medications, albumin infusion data, genetic analyses (where performed), any confirmed thrombo-embolic events, and any confirmed haemorrhagic events (both determined by clinical discussion). Estimated glomerular filtration rate (eGFR) was calculated using the Bedside IDMS-traceable Schwartz GFR equation (GFR (ml/min/1.73 m2) = (36.2 × length (cm))/creatinine (μmol/l)). In cases where length data was unavailable early in clinical course (n = 3), growth chart values were extrapolated backwards along their centile to provide an estimate of length at the time of presentation. The primary study endpoint was effective and stable thromboprophylaxis, defined as three consecutive therapeutic measurements. Therapeutic levels of enoxaparin were defined as anti-factor Xa levels of 0.2–0.4 IU/ml; therapeutic warfarinisation was defined as INR between 2.0 and 3.0. In patients where a thrombotic event occurred prior to anticoagulation, secondary thromboprophylaxis levels were targeted to anti-factor Xa levels of 0.5–1.0 IU/ml. Secondary endpoints were bilateral nephrectomies, transplantation, or the development of stage 5 chronic kidney disease (CKD 5), defined as confirmed eGFR < 15 ml/min/1.73 m2 (i.e., the value was calculated using a measured height, not via extrapolation). Where patients switched thromboprophylaxis modality, data were also collected from the onset of the second therapy, until the same endpoint was reached. Secondary outcomes included clinically confirmed VTE or any clinically significant episode of haemorrhage. Results Eleven children had a confirmed diagnosis of CNS between 1 July 2005 and 1 January 2018. Three children were not included. One child died at 2 weeks of age, one presented initially with severe acute kidney injury requiring haemofiltration and had a persistent requirement for dialysis thereafter for fluid removal (patient 9), and the third was in CKD 5 at the time of presentation (patient 10). Table 1 summarises the relevant demographic, phenotypic, and clinical details of all included patients. Supplementary Table 1 summarises excluded patients. There were five male patients and three female, with clinical presentation at a mean age of 6 weeks (range 2–15 weeks). Clinically, one patient had Pierson syndrome and two had Denys Drash syndrome. Histologically, four patients had diffuse mesangial sclerosis, two patients had ‘stage 5’ histological findings, one patient had mild glomerular change only, and one patient had no biopsy undertaken. Mutational analysis showed that five patients had mutations affecting NPHS1, one had a LAMB2 mutation, and two had WT1 mutations. Table 2 details the mutational analyses in patients where available. The eGFR at presentation was highly variable between patients (range 16–177 ml/min/1.73 m2) as was presenting serum albumin (range 6–21 g/L). Proteinuria data was available for 5/8 patients at presentation (range 3.81–9.63 g/mmol). Antithrombin III levels were measured in 2 patients at presentation, both below the normal range (patients: 25–61 IU/dL, normal: 71–101 IU/dL). Measurement of antithrombin III is not routine in our institution, and no other results at presentation were available.Table 1 Demographic and clinical summaries of all included patients Patient 1 2 3 4 5 6 7 8 Sex M M M M M F F F Associated phenotypic syndrome None None None None None Denys Drash Pierson Denys Drash Histology 50–80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, proximal tubular dilatation 80% global glomerulosclerosis, increased mesangial matrix, chronic interstitial inflammation, cystic tubular dilatation, marked interstitial fibrosis/tubular atrophy DMS 10% global glomerulosclerosis, 50% minor glomerular synechiae. Predominantly normal tubules. V mild interstitial fibrosis DMS DMS Not done DMS Genetic mutation (Table 2) NPHS1 homz NPHS1 comHet NPHS1 comHet NPHS1 comHet NPHS1 comHet WT1 LAMB2 WT1 Age at presentation (weeks) 3 2 2 9 4 15 7 2 Initial eGFR (ml/min/1.73 m2) 72 177 145 149 151 64 40 16 Initial Serum albumin (g/L) 11 10 6 10 6 13 21 6 Initial antithrombin III level (IU/dL) (normal 71-101) NM NM NM NM NM 25 61 NM Initial uPCR (g/mmol) NM NM 8.10 NM 3.81 6.96 8.83 9.63 Enoxaparin primary end point Never therapeutic, discontinued after 25 weeks 6 weeks to therapeutic Therapeutic at 6 weeks Never therapeutic after 27 weeks Therapeutic at 26 weeks CKD 5 at 10 weeks CKD 5 at 9 weeks Therapeutic at 3 weeks Warfarin primary end point 11 weeks to therapeutic 6 weeks to therapeutic N/A Never therapeutic after 50 weeks therapy Discontinued after 22 weeks due to bleeding concerns N/A N/A N/A Outcome Transplant aged 6 years Transplant aged 4 years Deceased (05/2020)—unknown cause Spontaneous improvement, now CKD3 aged 14 years Unilateral Nephrectomy Deceased aged 3 years Deceased aged 3 years Deceased aged 6 months Bilateral nephrectomy (06/2018), on PD Homz homozygous, comHet compound heterozygote, eGFR estimated glomerular filtration rate, uPCR urinary protein creatinine ratio, M male, F female, NPHS1 nephrin, LAMB2 beta-2-laminin, CKD 5 stage 5 chronic kidney disease, DMS diffuse mesangial sclerosis, NM not measured, PD peritoneal dialysis Table 2 Complete mutational analyses for all patients Patient Genetics 1 NPHS1: Homozygous mutation c.2417c > G Highly likely to be pathogenic 2 NPHS1: Compound heterozygote c.523C > T exon 5, nonsense c.1379G > A exon 11, missense Both highly likely pathogenic 3 NPHS1: Compound heterozygote c.1954C > T exon 15, nonsense c.2335-1G > A intron 17, skip/frameshift Likely pathogenic and highly likely pathogenic respectively 4 NPHS1: Compound heterozygote c.2335-1G > A intron 17 – skip/frameshift c.2491C>T exon 18 missense Highly likely pathogenic and likely pathogenic respectively 5 NPHS1: Compound heterozygote c.2227C > T exon 17 – missense c.2335-1G > A intron 17 – skip/frameshift Both classed highly likely pathogenic 6 WT1: Heterozygous c.[443-6C>A];[=] Classed as unlikely pathogenic 7 LAMB2: Homozygous splice site variant in intron 25 c.3982 + 1G > T Pathogenic, unknown effect but predicted to skip exon 25 8 WT1: De novo novel heterozygous frameshift variant on exon 9 c.[1201delA];[1202=] Likely pathogenic. 9 LAMB2: Homozygous c.736C > T exon 7 – missense Pathogenic 10 WT1: Heterozygous c.1181G > A exon 9 – missense NPHS1 nephrin, LAMB2 beta-2-laminin, WT1 Wilms tumour 1 All patients had a central venous catheter (CVC) inserted for either the delivery of intravenous albumin or the provision of haemodialysis. The albumin requirement varied from 6.3 to 31.5 g/kg/week. Further detail on albumin requirements are provided in Supplementary Table 2. Standard medical management in our unit also included regular administration of phenoxymethylpenicillin (penicillin V), levothyroxine as needed, angiotensin-converting enzyme inhibition (ACEi), and anti-reflux medications. Enoxaparin dosing All included patients were commenced on LMWH (enoxaparin) as a first-line thromboprophylaxis agent, at a mean starting dose of 1.88 mg/kg/day (range 0.71–4.3 mg/kg/day). The dose then subsequently varied from 0.71 mg/kg/day to a maximum of 7.44 mg/kg/day. All patients received subcutaneous administration twice a day with anti-factor Xa levels measured at 4 to 6 h post-dose. No patients received enoxaparin via infusion. Antithrombin III levels were not routinely measured, though 3 patients had at least one measurement (always below normal). No patient received antithrombin III infusions. Figure 1 details graphs of enoxaparin dosing, anti-factor Xa levels, eGFR, and serum albumin (Supplementary Figure 1 replaces serum albumin with urinary protein:creatinine ratio where available). Four patients reached therapeutic anti-factor Xa levels with the dose varying from 3.2 to 5.07 mg/kg/day. and time taken varying from 3 to 28 weeks (Table 1; patient 2 and 3: 6 weeks, 4.0 mg/kg/day and 5.07 mg/kg/day, respectively; patient 5: 26 weeks, 4.79 mg/kg/day; patient 8: 3 weeks, 1.82 mg/kg/day). Four patients did not reach therapeutic anti-factor Xa levels. Two patients reached CKD 5 before therapeutic levels were achieved, resulting in discontinuation of anticoagulation. Two patients had discontinuation due to failure to achieve adequate levels despite dose escalation, occurring after 25–27 weeks of therapy. The patients achieving therapeutic LMWH levels had NPHS1 compound heterozygote or WT1 mutations (patients 2, 3, and 5 = NPHS1 compound heterozygote, patient 8 = WT1 mutation). An apparent inverse relationship was noted between eGFR and anti-factor Xa levels, i.e., a decrease in eGFR associated with an increase in anti-factor Xa levels as might be physiologically expected. Serum albumin was proportional, with a higher serum albumin associated with higher anti-factor Xa levels.Fig. 1 Enoxaparin data. Graphs demonstrating individual patient enoxaparin dosing, therapeutic monitoring using anti-factor Xa, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays enoxaparin dose and anti-factor Xa level. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Warfarin dosing Four patients were subsequently commenced on warfarin, at a mean starting dose of 0.19 mg/kg/day (range 0.18–0.2 mg/kg/day). The dose then varied from 0.18 mg/kg/day to a maximum of 0.89 mg/kg/day. Figure 2 details graphs of warfarin dosing, INR, eGFR and serum albumin (Supplementary Figure 2 replaces serum albumin with uPCR for patient 5). Two patients reached therapeutic INRs with doses from 0.22 to 0.25 mg/kg/day and time taken varying from 6 to 11 weeks (Table 1; patient 1: 11 weeks, 0.22 mg/kg/day; patient 2: 6 weeks, 0.25 mg/kg/day). Two patients did not reach therapeutic INR. Patient 4 did not reach therapeutic levels after 1 year and patient 5 was discontinued from warfarin after 22 weeks due to concerns regarding bleeding. For eGFR and INR the graphs again show an inverse relationship.Fig. 2 Warfarin data. Graphs demonstrating individual patient warfarin dosing, therapeutic monitoring using INR, eGFR, and serum albumin. The left y-axis displays eGFR and serum albumin data; the right y-axis displays warfarin dose and INR. The grey shaded area represents the target therapeutic range for thromboprophylaxis. The vertical grey dotted line represents an adverse event Supplementary figure 3 provides similar information for non-included patients 9 and 10. Adverse events Tables 3 and 4 summarise identified adverse events in included patients (clinical vignette 1 provides the same for patient 9). Relevant kidney parameters and anticoagulation data at the time are included. Supplementary Table 3 details concomitant medications at the time of adverse events. There were two bleeding events and one thrombotic event during follow-up. One thrombotic event occurred prior to thromboprophylaxis in this cohort.Table 3 Anticoagulation and complication data for all included patients Patient 1st drug Starting dose (minimum-maximum) (mg/kg/day) Dose when therapeutic (mg/kg/day) Time to therapeutic dose eGFR start eGFR when therapeutic 2nd drug Starting dose (minimum–maximum) (mg/kg/day) Dose when therapeutic Time to therapeutic dose eGFR start eGFR when therapeutic Thrombus Bleeding 1 Enoxaparin 0.71 (0.71-5.14) N/A Never therapeutic 60.8 N/A Warfarin 0.19 (0.19–0.23) 0.22 11 weeks 36.4 59.6 N/A N/A 2 Enoxaparin 4.3 (2.9–5) 4.0 6 weeks 271.5 313.2 Warfarin 0.19 (0.19–0.25) 0.25 6 weeks 16.4 11.9 N/A N/A 3 Enoxaparin 2.3 (2.3-5.78) 5.07 6 weeks 145 150 N/A N/A N/A N/A N/A N/A N/A N/A 4 Enoxaparin 0.89 (0.89–5.62) N/A Never therapeutic 176.1 N/A Warfarin 0.2 (0.2–0.89) N/A Never therapeutic 295.5 N/A N/A N/A 5 Enoxaparin 1.9 (1.9–7.44) 4.79 26 weeks 226.25 145.9 Warfarin 0.18 (0.18–0.25) N/A Never therapeutic 93.1 N/A N/A 2 Bleeding events 6 Enoxaparin 2 (2–6.53) N/A Never Therapeutic 85.98 N/A N/A N/A N/A N/A N/A N/A Right femoral vein thrombus N/A 7 Enoxaparin 1.1 (1.1–6) N/A Never therapeutic 19.5 N/A N/A N/A N/A N/A N/A N/A N/A N/A 8 Enoxaparin 1.82 (1.82–3.48] 3.2 3 weeks 16.25 6.8 N/A N/A N/A N/A N/A N/A SVC thrombus pre-thromboprophylaxis N/A eGFR estimated glomerular filtration rate, N/A not applicable Table 4 Thrombotic and bleeding events and relevant parameters Patient Adverse event Age at event (weeks) Drug Time to event from starting medication (weeks) Dose (mg/kg/day) INR Anti-factor Xa level (IU/ml) eGFR (ml/min/1.73 m2) Serum albumin (g/L) Platelets (x 109/L) uPCR (g/mmol) Additional data 5 Bleeding 50 Warfarin 5 0.293 6 N/A 63.4 30 174 10.36 Blood altered vomiting and stools with infection in PEG 5 Bleeding 56 Warfarin 11 0.252 5.5 N/A 133.1 12 274 Nil Haematemesis with 1 week history of viral infection. Blood dried around gastrostomy site. 6 Thrombus – femoral vein 17 Enoxaparin 1 4.19 N/A 0.27 103.2 13 454 41.72 Haemodialysis dependent, low iron, hypothyroidism. 8 Thrombus – SVC 2 N/A N/A N/A N/A N/A 8 16 373 9.63 Managed in PICU, treated for maternal Grave’s disease eGFR estimated glomerular filtration rate, INR international normalised ratio, N/A not applicable Bleeding Patient 5 had two bleeding events after 5 and 11 weeks of therapy, both whilst on warfarin. This coincided with a supratherapeutic INR. The patient was haemodynamically stable on both occasions. The first bleeding event occurred 3 months following unilateral nephrectomy, whilst on home IV albumin. The patient presented with fresh red blood evident in the stool, with visible clot. The patient’s gastrostomy was noted to be leaking with evidence of superficial infection. Indomethacin was temporarily discontinued, IV omeprazole administered, and warfarin withheld. The INR was 6. Packed red cells were transfused to improve haemoglobin (pre-transfusion, 54 g/L). Twelve hours post-presentation, there was fresh blood leakage from the gastrostomy, coinciding with coffee-ground vomiting. IV vitamin K was administered at a dose of 30 mg/kg to reverse over-warfarinisation without preventing ongoing thromboprophylaxis. Warfarin was withheld for 48 h then re-commenced at the original dose. The second bleeding event occurred 1 week following an upper respiratory tract infection, 1 month after the initial bleeding event, presenting again with blood-specked vomitus and fresh blood leakage from the gastrostomy. Haemoglobin had fallen from 99 to 70 g/L. INR was ‘unrecordable’ twice, so IV vitamin K was administered, again at 30 mg/kg. Repeat INR 6 h later was 5.5. Transfusion was not required on this occasion. Warfarin was recommenced at a slightly lower dose after 72 h. Two months later, the same patient then had an incidental finding of an INR of 8.8 with no associated bleeding symptoms. At that point, warfarin was discontinued and the patient re-commenced on LMWH. Thrombus No thrombotic complications developed whilst patients were adequately warfarinised. Patient 6 had identification of a femoral vein thrombus aged 4 months, 2 weeks following initial presentation. Initial management required continuous veno-venous haemofiltration (CVVH) initially via a femoral CVC, which was changed to a left internal jugular CVC 3 days into therapy. CVVH was discontinued after 4 days, and the patient was commenced on enoxaparin. One week later, the patient developed evident discrepancy in leg size, with identification of non-occlusive thrombus within the right femoral vein. This coincided with a thromboprophylactic anti-factor Xa level of 0.27 IU/ml. At the time of thrombus detection, the patient was proteinuric (uPCR of 41.72 g/mmol), hypoalbuminaemic (13 g/L), and had a mild thrombocytosis (454 × 109/L). Following detection of the thrombus, the target anti-factor Xa was temporarily increased to 0.5–1.0 IU/ml until the clot resolved, and for 3 months subsequently. Patient 8 developed a superior vena cava (SVC) thrombus 5 days following initial insertion of an internal jugular CVC at 2 weeks of age, prior to the commencement of anticoagulation. Enoxaparin was subsequently initiated as secondary thromboprophylaxis, with target levels of 0.5–1.0 IU/ml. Of note, the patients’ mother also had Grave’s disease, which may have further exacerbated thrombosis risk. At the time of database lock, two patients had successfully been transplanted, four patients had died (cause of mortality: sepsis = 1, cardiomyopathy = 1, intestinal obstruction and perforation = 1, probable autonomic failure = 1), one patient was on peritoneal dialysis, and one had ongoing CKD stage 3. Discussion This case series describes the challenges in achieving effective and safe thromboprophylaxis in patients with CNS. Enoxaparin led to adequate thromboprophylaxis in 4/8 patients compared with 2/4 patients on warfarin, with variable therapeutic times and doses. Both agents had similar safety profiles. All bleeding complications were associated with supra-therapeutic measurements, highlighting the requirement for careful monitoring. Anti-factor Xa levels and INR appear to have an inverse relationship with kidney function, as might be physiologically expected. Loss of kidney function reduces proteinuric losses of antithrombin III and other relevant proteins, which may contribute to more effective anticoagulation. The British National Formulary for children (BNFc) is the standard formulary within the UK and recommends an initial enoxaparin dose of 1 mg/kg/day for secondary thromboprophylaxis for children aged over 2 months (an initial dose of 2 mg/kg/day is recommended under 2 months, due to differences in infant drug handling) [23]. International guidelines suggest higher doses for younger children [14]. Our study cohort all received higher doses than BNFc guidelines, both initially and once therapeutic. The mean initial dose in our cohort was 1.88 mg/kg/day, nearly double the recommended starting dose, with the therapeutic dose ranging from 3.2 to 5.07 mg/kg/day. The mean enoxaparin dose required to achieve adequate primary thromboprophylaxis was 4.27 mg/kg/day, over 4 times the suggested dose. The requirement for higher doses may be attributable to a generally younger age, lower antithrombin III levels related to proteinuric loss (below the normal range in all patients where measurement was performed; Table 1), and potentially other relevant urinary losses [14, 18]. Dosing variability likely also reflects the genotypic and phenotypic differences within our small cohort, including the degree of proteinuria. Though therapeutic monitoring is not generally undertaken in adults on enoxaparin, the volatile nature of both proteinuria and kidney function mandates monitoring in paediatric patients. All patients in this cohort had administration of enoxaparin twice daily, though once daily dosing is also described. Though there are no reported differences in safety or efficacy between a once or twice daily dosing regimen, the available pharmacokinetic data supports a twice daily dosing regimen [24, 25]. As expected, warfarin dosing was variable between patients and required careful titration and monitoring, similar to other patient groups. Our cohort’s mean initial dose was 0.19 mg/kg, similar to the recommended initial dose of 0.2 mg/kg. Our cohort reflects the known literature, with warfarin dosing ranging from 0.18 to 0.89 mg/kg, and a mean dose of 0.24 mg/kg achieving an INR suitable for primary thromboprophylaxis. In one prospective study, infants required higher doses of warfarin than older children, with infants under 1 requiring ~ 0.32 mg/kg, whereas children over 11 years required ~ 0.09 mg/kg [20]. Patient 4 never reached a therapeutic INR despite dose escalation to 0.89 mg/kg. Warfarinisation of children is challenging, even more so in patients with ongoing alterations in their haematologic physiology [16, 21]. To our knowledge this is the first study to address and report actual monitoring of thromboprophylaxis in a national cohort of CNS patients. A recent multi-centre retrospective review of anti-thrombotic prophylaxis was carried out in 17 centres over 15 European countries. The investigators reported that 4/45 (11%) receiving anticoagulants and 5/26 (15%) not receiving anticoagulants developed VTEs (p = 0.60). Notably, the majority of VTEs in that cohort occurred whilst patients were warfarinised (warfarin in 3, heparin in 1, aspirin in 1). This finding contrasts with our observation of VTEs only occurring in a heparinised patient, though our cohort is both smaller and has a different genetic mix (69% NPHS1 and 14% WT1 in Dufek et al., 50% and 25% respectively for our cohort) [22]. A separate retrospective review of anticoagulated CNS patients reported a VTE rate of 29% (16/55). About 67% (37/55) of that cohort had an NPHS1 mutation, and no patients had a LAMB2 mutation—unlike the 2/8 in our cohort [11]. Our cohort has a relatively high prevalence of non-NPHS1 mutations or novel NPHS1 mutations, which may limit the comparability and generalisation of our results. Neither of the two larger studies reported assays indicating effective thromboprophylaxis, or whether dosing and kidney function influenced anticoagulant efficacy. Two further retrospective studies have investigated prophylactic anticoagulation in adults with nephrotic syndrome (NS). A Danish retrospective analysis investigated 79 patients; of whom 44 were anticoagulated and 35 were not and reported a significant reduction in thrombotic events (4 versus 0 episodes, p = 0.035) in patients receiving anticoagulant therapy without increasing bleeding episodes (p = 0.45) [26]. A second retrospective study reported thrombotic events in 1.39% (2/143) of anticoagulated patients and concluded that anticoagulation effectively reduced the VTE rate in nephrotic syndrome which reportedly ranges from 7 to 40% [27]. Though the adult NS literature suggests a role for thromboprophylaxis in reducing the VTE risk, the aetiology of adult NS is very different, even to idiopathic childhood NS, which is a further separate clinicopathological entity to CNS, including the degree of proteinuria which is typically many fold higher in CNS than idiopathic NS. Extrapolating findings from adult studies to this patient cohort must be done with caution. Within our cohort, only 50% (4/8) of heparinised and 50% (2/4) of warfarinised patients achieved adequate thromboprophylactic levels prior to the onset of CKD 5. Bleeding events occurred in 1 of 4 warfarinised patients. The only thrombosis on treatment developed with enoxaparin at an adequate thromboprophylactic level. The small sample size precludes formal analysis or recommending one agent over another. All patients were initially heparinised, with warfarin used as second-line thromboprophylaxis in our unit. It is plausible that adequate thromboprophylaxis is more readily achieved later in the disease course, due to patients being more stable, or having reduced overall proteinuric loss. A larger cohort of patients receiving either warfarin or enoxaparin initially would be required to truly determine the more efficacious agent. For reasons previously described, this is unlikely to occur. Patient 7 required a significantly lower dose of enoxaparin to reach target anti-factor Xa levels. This could be partly explained by the patient’s early development of significant CKD and lesser degree of proteinuria. This patient also represents the only included patient with LAMB2 mutation, again indicating genotypic variability. All patients had CVCs. This is an established risk factor for the development of VTEs; in one reported cohort ~ 5% of paediatric patients with CVCs in situ had at least one VTE [28]. In both cases of thrombus in this cohort (patient 6 and 8), thrombus was detected within a catheterised or recently catheterised vessel, and within 2 weeks of initial presentation. As a CVC is often fundamental to CNS management, risk mitigation can only be via timely thromboprophylaxis. Using higher than BNFc recommended initial dosing may achieve this, though that conclusion cannot be drawn from our cohort [14]. Warfarin has many potential medication interactions which could have prevented target INRs. All warfarinised patients were prescribed antibiotics concurrently which could have altered warfarin’s pharmacodynamics. Additionally, patient 5 developed a central line sepsis and thrombocytopenia. This could partly explain why this patient had repeated bleeding events coinciding with supraphysiological INRs. Yet, in this patient population there are likely to be many unavoidable confounders to therapeutic warfarinisation due to the complexities of CNS management. Though multiple medications can potentiate or inhibit the actions of thromboprophylaxis, the doses of concomitant medications used routinely in these patients (e.g. antibiotic prophylaxis) were typically standard and infrequently altered. The effect on thromboprophylaxis pharmacokinetics would therefore be consistent and unlikely to account for sudden changes in INR or anti-factor Xa. These patients are complex with multiple factors impacting on both pharmacokinetics and pharmacodynamics—further supporting the need for regular therapeutic surveillance. The management of CNS typically includes regular infusions of IV albumin, the dose of which reflects the degree of proteinuria. Weekly albumin doses varied within the cohort from 5 to 32 g/kg/week (Supplementary Table 2). There was no apparent association between dose of albumin administered and likelihood of achieving adequate thromboprophylaxis. Patient 4 in this cohort never required IV albumin, and had a different clinical course, similar to that seen in Maori populations. Yet this patient was the most difficult patient to manage thrombotic risk, failing both LMWH and warfarin despite prolonged treatment with both [1]. Two patients had a long period of sub-therapeutic treatment of enoxaparin with minimal dosing changes (Fig. 1: patient 1: 25 weeks, patient 2: 27 weeks). Prolonged sub-therapeutic therapy could increase the VTE risk, necessitating consideration of conversion to warfarin. Achieving effective thromboprophylaxis for these patients was challenging, as in some eGFR increased with time, possibly resulting in elevated clotting factor excretion. Clinical instability may cause clinicians to be reluctant to alter medication dosage, which may partly explain the long sub-therapeutic period. Conversely, one warfarinised patient was converted back to enoxaparin due to safety concerns from unstable and excessive INR, and two episodes of gastrointestinal bleeding. The cohort is from a single national centre with 100% patient identification over a 15-year period, with all patients treated by the same clinical team thereby reducing variability in clinical treatment. This dataset is (to our knowledge) unique in showing the relationship between anticoagulant dosing, therapeutic drug levels, and kidney function in patients with CNS. The optimal therapeutic regimen in this patient population has not been ascertained. Though our cohort is too small to definitively comment on dosing regimen or choice of thromboprophylaxis, the safety profiles confirm the importance of measuring therapeutic levels regularly in this complex patient group. There are limitations to this cohort. The patient group were heterogeneous, histologically and genetically, which may have conferred different risk profiles of VTE [27]. The variability in clinical course affecting both proteinuria and kidney function will also have an impact on interpretation. This heterogeneity further highlights the difficulties in establishing an evidence base for thromboprophylaxis in CNS. The small sample size precludes statistical analysis, unavoidable due to the disease rarity. A sufficiently large cohort would mandate further international trials, but the most recent effort demonstrated how challenging this is. Despite engaging 22 tertiary European centres, that study failed to recruit enough patients to achieve statistical power for outcomes [22]. The limited data on proteinuria prevents interrogation of the relationship between therapeutic drug levels and urinary protein. Retrospective review of healthcare records for outcome reporting is recognised to have flaws, as minor but clinically relevant episodes may not be reported or poorly documented. This is somewhat mitigated by the lengthy in-patient stays of these patients. All adverse events have occurred in a hospital setting. For three patients (4–6) length data was unavailable in the early parts of life, so eGFR was calculated by retrospective extrapolation using the patient’s nearest available length centile. This may overestimate earlier length as early management of CNS includes optimising nutrition and growth. To limit the impact of this, the outcome of CKD 5 was only assigned when using either a confirmed patient length, or where kidney replacement therapy was required. It is plausible that early kidney function was overestimated for those patients. Conclusions This case series demonstrates that achieving adequate and stable thromboprophylaxis in children with CNS is challenging. All bleeding events were associated with supra-therapeutic levels. Development of thrombus prior to or shortly after any thromboprophylaxis highlights the importance of commencing this early. Enoxaparin doses required for thromboprophylaxis in this patient population were approximately double the recommended dose. Electronic supplementary materials ESM 1 (DOCX 233 kb). Abbreviations BNFc British National Formulary for Children CNS Congenital Nephrotic Syndrome CVVH Continuous veno-venous hemofiltration eGFR Estimated glomerular filtration rate INR International Normalised Ratio LMWH Low molecular weight heparin SVC Superior vena cava VTE Venous Thromboembolism UPCR Urinary protein:creatinine ratio Acknowledgements Thanks to Rowan Davis and Robin Oswald for involvement in data collection, to the clinical teams caring for these patients, and the families themselves. Authors’ contributions All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by LJD, AL, LE and BCR. AL, BCR and IJR had clinical oversight of all included patients. The first draft of the manuscript was written by LJD, and all authors commented on subsequent versions of the manuscript. All authors read and approved the final manuscript. BCR serves as the data guarantor. Data availability The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Compliance with ethical standards Conflicts of interest The authors declare that they have no conflict of interest. Ethical approval This study was a review of clinical management so ethical approval was not required. Every investigator involved in the initial review of patient records was an approved healthcare provider for these patients, and so chart review was undertaken by the clinical treating team. Consent to participate Families were consented clinically; data was suitably anonymised. Consent for publication Families were consented clinically; data was suitably anonymised. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Recovered
ReactionOutcome
CC BY
33089377
19,416,137
2021-05
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Hyperlactacidaemia'.
Prevalence and risk factors of lactic acidosis in children with acute moderate and severe asthma, a prospective observational study. Lactic acidosis is a common complication of status asthmaticus in adults. However, data is sparse in children. The aim of this study was to describe the prevalence and risk factors for lactic acidosis in children hospitalised for acute moderate or severe asthma. A total of 154 children 2-17 years of age were enrolled in a prospective observational study conducted in a tertiary hospital. All had capillary blood gas assessment 4 h after the first dose of salbutamol in hospital. The primary endpoint was the prevalence of lactic acidosis. Potential contributing factors such as age, sex, BMI, initial degree of asthma severity, type of salbutamol administration (nebuliser or inhaler), steroids, ipratropium bromide, and glucose-containing maintenance fluid represented secondary endpoints. All in all, 87% of patients had hyperlactatemia (lactate concentration > 2.2 mmol/l). Lactic acidosis (lactate concentration > 5 mmol/l and anion gap ≥ 16 mmol/l) was observed in 26%. In multivariate analysis, age more than 6 years (OR = 2.8, 95% CI 1.2-6.6), glycemia above 11 mmol/l (OR = 3.2 95% CI 1.4-7.4), and salbutamol administered by nebuliser (OR = 10, 95% CI 2.7-47) were identified as risk factors for lactic acidosis in children with moderate or severe asthma.Conclusion: Lactic acidosis is a frequent and early complication of acute moderate or severe asthma in children. What is Known: • Lactic acidosis during acute asthma is associated with b2-mimetics administration. • Salbutamol-related lactic acidosis is self-limited but important to recognise, as compensatory hyperventilation of lactic acidosis can be mistaken for respiratory worsening and lead to inappropriate supplemental bronchodilator administration. What is New: • Lactic acidosis is a frequent complication of acute asthma in the paediatric population. • Age older than 6 years, hyperglycaemia, and nebulised salbutamol are risk factors for lactic acidosis during asthma. Introduction Asthma is characterised by chronic airway inflammation and hyperresponsiveness. During an acute exacerbation, inhomogeneous airway narrowing and obstruction lead to hypoxemia. Compensatory hyperventilation mediated by pulmonary mechanoreceptors conducts to respiratory alkalosis, the most frequent acid-base disturbance observed in acute asthma. If not treated properly, progressive respiratory insufficiency may occur with hypercapnia and respiratory acidosis [1]. Lactic acidosis is another blood gas alteration observed during moderate or severe asthma. Multiple mechanisms have been proposed. Some have evoked reduced tissue perfusion or overuse of respiratory muscles under hypoxic conditions [2]. Others have suggested that b2-adrenergic agents like bronchodilators used to treat asthma lead to increased gluconeogenesis, glycogenolysis, glycolysis, and lipolysis, cumulating in lactic acid production [3–6]. Depending on the mechanism of lactate formation, two types of lactic acidosis exist that can be distinguished calculating the lactate to pyruvate ratio (L/P). Type A (L/P ratio < 25/1) is related to impaired oxygenation, and type B (L/P > 25/1) is caused by excessive b2-receptor stimulation [6]. Lactic acidosis is now a well-known complication of status asthmaticus in adults. Case reports and some retrospective and rare prospective studies describe a transient lactic acidosis as a side effect of high doses b2-agonists used in acute asthma treatment. However, data is rare in children [7]. Even if lactic acidosis during asthma is a self-limited condition, it has an impact on assessment and management of respiratory distress. Compensatory hyperventilation of lactic acidosis is often mistaken as a sign of respiratory worsening and leads to inappropriate escalation of bronchodilator therapy, increasing morbidity and mortality [8, 9]. The aim of our study is to describe the prevalence and risk factors contributing to lactic acidosis in children treated with salbutamol for moderate or severe acute asthma. Materials and methods Study design This prospective observational monocentric prevalence study was conducted from May 01, 2017, to April 30, 2019, in a tertiary care children’s hospital. Patients Children and adolescents 2 to 17 years of age hospitalised for acute moderate or severe asthma were eligible for the study. At our institution, patients requiring inhaled b2-agonists minimum every 2 h fill the indications to be admitted, and an initial degree of asthma severity based on PRAM score is part of the clinical evaluation [10]. Exclusion criteria were as follows: parent’s refusal, metabolic disorder, shock, sepsis, renal or hepatic insufficiency, diabetes mellitus, and cancer. Data collection Basic clinical data included age; body mass index (BMI); sex; initial degree of asthma severity; dose and type of salbutamol administration, of oral or intravenous steroids, and of inhaled ipratropium bromide; and type of maintenance intravenous fluid. Hypoxemia (defined as SpO2 < 92%) was rigorously assessed and treated. Salbutamol was administered by using a metered dose inhaler (pMDI) (through a valved holding chamber with a mouthpiece or a mask, when necessary, AeroChamber Plus Flow Vu®, 1push = 100 cmg of salbutamol) or nebuliser (aerosol solution of 5 mg of salbutamol in 5 ml of normal saline solution NaCl 0.9%). Three types of maintenance fluid were used: 91% of G10% and 9% of NaCl 10% mix, 91% of G5% and 9% of NaCl 10% mix, or normal saline solution (NaCl 0.9%). Capillary blood sample was drawn 4 h after the first administration of salbutamol in hospital, or in case of secondary appearance of tachypnoea or worsening of the respiratory status during hospitalisation. Oxygen saturation was measured by pulse oximetry before blood extraction. Blood gas assessment including the measurement of pH, pCO2, HCO3, base excess (BE), and glucose and lactate concentrations was performed on a RAPIDPoint500, Siemens, gasometer. One millilitre of blood was used for analysis. Hyperlactatemia was defined as lactate concentration > 2.2 mmol/l. Lactic acidosis was defined as lactate > 5 mmol/l and anion gap (AG) ≥ 16mmol/l (AG = Na + K-HC03-Cl), non-compensated lactic acidosis as pH < 7.35, and lactate concentration as > 5 mmol/l. Compensated lactic acidosis represented lactate concentration > 5 mmol/l, pH ≥ 7.35, and pCO2 < 35 mmHg. Hyperglycaemia was characterised as glucose > 11 mmol/l. Outcomes The primary outcome was the prevalence of lactic acidosis. Secondary outcomes included other contributing factors like age, sex, BMI, initial degree of severity, salbutamol administered by inhaler or by nebuliser, steroids, ipratropium bromide, and glucose-containing maintenance fluid. Ethical considerations Patients older than 11 years and parents or legal guardians were informed orally and in writing about the research project by one of the doctors working in the paediatric emergency room during admission to the hospital. Adolescents ≥ 14 years and their parents or legal guardians provided informed consent. The study was approved by the institutional ethics committee (Swiss Ethics, protocol number 2016-01320). Statistical analysis Categorical data were described as absolute counts. Percentages and continuous data we described as means and standard deviations (SD). The Mann–Whitney U test was used to compare the means. We firstly realised simple logistic regression and calculated odd ratio for all potential risk factors. All variables potentially able to influence lactic acidosis (p < 0.2) were used as covariates in multiple logistic regression. Pearson’s correlation coefficient was used to measure statistical relationship between the levels of lactates and doses of salbutamol. All statistical analysis was performed with the Epi Info version 7.2.3.1 software (Centres for Disease Control and Prevention). Results Among 627 patients from 2 to 17 years of age hospitalised for moderate or severe asthma, 174 received information about the study. Eleven did not provided informed consent, and 9 did not have blood sample for technical problem. Finally, 154 patients were included in the study (Fig. 1)Fig. 1 Flow chart of study population Among 154 patients, 99 (64%) were aged from 2 to 6 years and 55 (36%) were more than 6. A total of 13% were obese (P > 97‰). Sex ratio was 1.92 male to female. Forty-three percent of episodes were categorised as severe (PRAM > 8) and 57% as moderate (PRAM 4–8). Salbutamol was administered by pMDI with holding chamber alone in 30% of cases (46/154) or by nebuliser (alone or associated with pMDI administration) in 70% (108/154) (Table 1). Mean overall dose of salbutamol administered during the first 4 h after hospital admission was 12 ± 10 mg. The mean dose was similar in older (> 6 years of age) and younger groups (≤ 6 years of age), 12.3 ± 10.5 mg and 11.8 ± 9.8 mg respectively (p = 0.59). Patients with severe asthma received higher doses of salbutamol (mean 19 ± 10.1 mg) compared to patients with moderate asthma (mean 6.67 ± 5.7 mg), p < 0.0005. When delivered by nebulisation (1 nebulisation = 5 mg of salbutamol), the mean dose of salbutamol was 16 ± 9.3 mg versus 2.4 ± 1.2 mg when delivered by inhaler (1 push = 100 cmg), p < 0.0005.Table 1 Characteristics of study population Characteristics n = 154 % Age ≥ 6 years of age 55 36 Female sex 51 33 Severe asthma (PRAM 8–12) 66 43 Moderate asthma (PRAM 4–7) 88 57 PICU admission 0 0 Obesity 20 13 Salbutamol administered by inhaler 46 30 Salbutamol administered by nebuliser 108 70 Intravenous salbutamol 0 0 Aminophylline 0 0 Corticoids 146 95 Ipratropium bromide 23 15 Glucose-containing maintenance fluid 30 19 Hyperlactatemia > 2.2 mmol/l 134 87 Hyperlactatemia > 5 mmol/l 40 26 Non-compensated lactic acidosis (lactate > 5 mmol/l and pH < 7.35 mmHg) 6 4 Compensated lactic acidosis (lactate > 5, pH > 7.35, pCO2 < 35 mmHg) 34 22 Hyperglycaemia 56 36 Almost all patients (95%) received corticosteroids with a mean dose of 1.85 ± 0.75 mg/kg. Corticosteroids were mainly administered orally (88%). Twenty-three of 154 patients (15%) received ipratropium bromide. Nineteen percent were perfused with glucose-containing maintenance fluid (91% of G10% and 9% of NaCl 10% mix or 91% of G5% and 9% of NaCl 10%) (Table 1). All patients had capillary blood gas analysis 4 h after the first dose of salbutamol administration. Secondary appearance of tachypnoea or worsening of the respiratory status motivated a second blood gas analysis in 13 patients. Most of the patients (87%) had mild hyperlactatemia (lactate > 2.2 mmol/l). All of patients with lactate concentration > 5 mmol/l (26%) had lactic acidosis (lactate > 5 mmol/l and augmented anion gap AG ≥ 16 mmol/l). Thirty-four (22%) presented compensated lactic acidosis (lactate > 5 mmol/l, pH ≥ 7.35, and pCO2 < 35 mmHg). Only 6 (4%) had pH < 7.35. None had hypercapnia (pCO2 > 40 mmHg) (Table 1). In univariate analyses, a significant correlation was found between lactic acidosis and female sex (OR = 2, 95% CI 1–4.2), as well as between lactic acidosis and severe asthma (OR = 4, 95% CI 1.9–8.6). When adjusted on potential confusing factors in multivariate analysis, salbutamol administered by nebuliser (aOR = 10, 95% CI 2.7–47), age older than 6 years (aOR = 2.8, 95% CI 1.2–6.6), and hyperglycaemia (aOR = 3.2 95% CI 1.4–7.4) were related to increased risk of lactic acidosis (Table 2).Table 2 Risk factors of lactic acidosis Risk factors of lactic acidosis Number of patients (%) Univariate analysis unadjusted OR [95% CI] Multivariate analysis adjusted OR [95% CI] Lactic acidosis (yes/non) n = 40 n = 114 Female sex 18 (45) 33 (29) 2 [1–4.2] 1.9 [0.8–4.4] Age ≥ 6 years of age 19 (48) 36 (32) 1.9 [1–4.1] 2.8* [1.2–6.6] Severe asthma 27 (68) 39 (34) 4 [1.9–8.6] 2 [0.8–5.2] Obesity 8 (20) 12 (11) 2.1 [0.8–5.7] 2.5 [0.8–8.1] Hyperglycemia (> 11 mmol/l) 24 (60) 32 (28) 3.8 [1.8–8.2] 3.2* [1.4–7.4] Glucose-containing maintenance fluid 8 (20) 22 (19) 1.05 [0.4–2.6] Corticoids 41 (100) 106 (95) Ipratropium bromure 8 (20) 15 (13) 1.7 [0.6–4.3] Salbutamol administered by nebulisation 38 (95) 70 (61) 11.9 [2.7–52] 10* [2.3–47] OR, odds ratio; CI, confidence interval Number of patients was expressed in absolute number and in percentage (%) All variables associated with p < 0.2 were included in the multivariate analysis Significative findings were marked with an asterisk (*p < 0.05) Lactic acidosis was observed even with low doses of salbutamol (< 0.5 mg/kg). No correlation was found between lactate levels and salbutamol doses (Pearson’s r = 0.17 (SE = 0.24)) (Fig. 2).Fig. 2 Relation between lactate levels and doses of salbutamol Discussion Although lactic acidosis is a well-known metabolic disturbance of asthma in adults, data are rare in the paediatric population. To our knowledge, this is the first large prospective study describing prevalence and risk factors for lactic acidosis in children with acute moderate or severe asthma. A total of 87% (134/154) of our patients had increased blood lactate concentration (lactate > 2.2 mmol/l) 4 h after the first salbutamol dose in hospital. Our findings compare to those of Meert who recorded 83% (62/105) of children with mild hyperlactatemia (lactate> 2.2 mmol/l) during status asthmaticus [5]. Lower prevalence (71%) was found in a retrospective study of 75 children, but the time of lactatemia assessment was not specified [11]. In adults, the prevalence of mild hyperlactatemia (lactate > 2 mmol/l) ranges from 59 in 29 patients with severe asthma 4 to 6 h after therapy beginning [12] to 69.2% at an earlier time point for lactate measurement (about 1 h 25 min after albuterol treatment beginning) [13]. In paediatric case reports, lactate concentration in asthma-related lactic acidosis ranged from 5.9 to 9.2 mmol/l [8]. Koul documented lactic acidosis with a peak lactate range (5.2–13 mmol/l) 2–8 h after the beginning of aerosol therapy in 4 children 11 to 17 years of age. In our study, lactatemia varied from 1.4 to 9.66 mmol/l 4 h after salbutamol administration, with 26% of patients presenting respiratory-compensated lactic acidosis (lactate > 5 mmol/l and AG ≥ 16 mmol/l, pCO2 < 35 mmHg), consistent with other observations [14]. In a retrospective study of 75 children with acute asthma, metabolic acidosis (pH < 7.35 and BE < − 7) was found in 21% of patients [11]. Available lactate level was > 5 mmol/l in 22% of children. Likewise, Meert identified 28% of 53 patients with metabolic acidosis (pH < 7.35, PCO2 < 35 mmHg, and BE < − 7 mmol/l) during acute asthma, with lactate assessment from 7.2 to 9.3 mmol/l 8 to 24 h after admission [4]. Lastly, lactic acidosis with or without respiratory compensation was identified in 47 of 105 (45%) children with acute asthma [5]. In critically ill paediatric patients with asthma, acidosis (pH < 7.35) was found in 45% of patients admitted to the intensive care unit (ICU). Only one had metabolic acidosis with hyperlactatemia (4.6 mmol/l 6 h after ICU admission). All the others had acidosis from respiratory origin. However, in these patients, the blood gas determination was realised quite early, within 2 h after emergency room admission, and the dose of salbutamol received was not specified. Yousef did not find metabolic acidosis in eight other episodes of severe respiratory failure attributable to asthma and suggested that lactic acidosis during asthma is not underestimated and children may be more resistant than adults to the development of this complication. He implied that metabolic acidosis reported in previous studies could be rather related to ketosis following suboptimal hydration and caloric management [15]. We cannot support this hypothesis because all our patients unable to feed or to drink received intravenous glucose perfusion. Lactic acidosis implies two mechanisms. Type A is associated with impaired oxygen delivery and/or hypotension. Type B implies underlying disease (liver or renal insufficiency, diabetes mellitus, or cancer), drugs (such as b2-agonists), or inborn errors of metabolism [16]. None of our patients had known chronic underlying disease. Many authors thought lactic acidosis during asthma to be type B [4–6]. Exposition to high doses of bronchodilator-type salbutamol induces hyperadrenergic state and leads to increased lactate production. Presence of lactic acidosis in patients receiving b-2 agonist therapy under optimal oxygenation or artificial ventilation supports this hypothesis [17]. On the biological level, types A and B can be distinguished by the L/P ratio (L/P < 25/1 versus L/ P> 25/1, respectively). Meert calculated the L/P ratio and concluded that type B lactic acidosis is the most frequent in asthma [5]. Even if hyperlactatemia has been described as a marker of mortality in critically ill patients [18], type B lactic acidosis is a self-limiting condition, and no fatal case has been described in children. The spontaneous resolution with decreasing doses or discontinuation of bronchodilator therapy is a rule [4, 5]. In our study, we did not perform pyruvate assessment for technical reasons and thus could not ascertain the mechanism of lactic acidosis precisely, but we highly suggest type B because none was hypoxemic at the time of lactate assessment and a favourable evolution was observed for all. In our patients, mean total dose of salbutamol delivered by nebulisation (16 ± 9.3 mg) was almost seven times higher compared to the mean dose delivered by inhalator (2.4 ± 1.2 mg). Higher doses could explain the greater risk of lactic acidosis if salbutamol is administered by nebulisation (RR = 10, 95% CI 2.3–47). According the Cochrane database, other side effects of salbutamol such as increased pulse rate were lower for pMDI in children (mean difference − 5% baseline, 95% CI − 8 to − 2%), as was the risk of developing tremor (RR = 0.64; 95% CI 0.44 to 0.95) [19]. On the other hand, in the paediatric population, only 1–10% nebulised salbutamol reaches the inferior respiratory tract [20–23]. Previous study by Wildhaber has shown equivalent percentages of total lung deposition of radiolabeled salbutamol aerosolised by using either a nebuliser or a pMDI with holding chamber (9.6% and 11% for inhaled and nebulised respectively in children > 4 years of age and 5.4% for both in children < 4 years of age) [22]. In a more recent study, it was shown that the amount of drug delivered from pMDI was higher, ranging from 18.1 to 22.5% in young children (3–5 years of age) [22, 23] and from 35.4 to 54.9% in older children (5–17 years of age) [24]. The authors concluded that most children from 5 years of age could obtain lung deposition of more than 30% using a tidal breathing technique with a pMDI. All our patients receiving salbutamol by inhaler via pMDI used this inhalation technique. We did not find a correlation between lactate levels and doses of salbutamol. The statistical power of a dose correlation would be probably reduced by a large proportion of children with mild hyperlactatemia (2.2–5 mmol/l). Intravenous, oral, and inhaled salbutamol raise glycogenolysis resulting in hyperglycaemia. Concurrent use of corticosteroids may exacerbate blood glucose level [6]. In our study, almost all patients received concomitant steroids (95%), and 36% of them had hyperglycaemia (glucose > 11 mmol/l). b2-agonist drugs have two main actions. Firstly, b2-adrenergic receptor stimulation increases glycogenolysis, neoglucogenesis, and glycolysis leading to transformation of glucose to glucose 6-phosphate and then to pyruvate. Secondly, b2-agonists enhance lipolysis. Free acids inhibit pyruvate dehydrogenase, an enzyme which normally allows pyruvate to enter the Krebs cycle. In this way, pyruvate to lactate formation is promoted [25]. In our study, we show that hyperglycaemia raises the risk of lactic acidosis during asthma (aOR = 3.2 95% CI 1.4–7.4). On the intracellular level, the rise in glucose blood level via bronchodilator-mediated glycolysis provides more substrate for lactate production. In patients with severe asthma in ICU, serum glucose was measured. Even if 88% of them had hyperglycaemia (> 6.8 mmol/l), the relationship between hyperglycaemia and lactate concentration could not be proved [4]. Our study showed that children aged more than 6 have almost three times more risk to develop lactic acidosis during asthma (aOR = 2.8, 95% CI 1.2–6.6), despite the fact that the mean dose of salbutamol was the same in both age groups (mean of 11.8 mg, SD 9.8 for < 6 years versus 12.3 mg, SD 10.5 for > 6 years). It could be related to the fact that younger children dispose less glucose resources and in consequence less substrate for lactate production. We identified other parameters like female sex, severe asthma, and obesity as independent risk factors for lactic acidosis. Even if they could not be confirmed in multivariate analysis, they need to be put forward as our study is the first one to try to identify potential risk factors of lactic acidosis in children with asthma. Our study has some limits. Firstly, only 25% of potential patients with moderate or severe asthma seen in the emergency room were included, which is a major selection bias. Secondly, we performed only capillary lactate assessment. However, prior research suggests that capillary lactate value accurately reflects arterial lactate [26]. Moreover, this technique is quicker and easier to perform, especially in children. Another limitation is the lack of initial lactate level. It would be interesting to compare the lactate level upon arrival and 4 h later. Finally, the relationship between steroid therapy and lactic acidosis could not be investigated because almost all patient received corticosteroids. Conclusion Lactic acidosis is a frequent and early (H4) complication of asthma observed in children treated with high doses of bronchodilators. Salbutamol administered by nebuliser, age more than 6 years, and hyperglycaemia were identified as risk factors of lactic acidosis during asthma. Even if self-limited, this condition is important to recognise to avoid unnecessary and harmful therapeutic intensification. Abbreviations AG Anion gap aOR Adjusted odds ratio BE Base excess BMI Body mass index CI Confidence interval L/P Lactate to pyruvate ratio OR Odds ratio pMDI Pressurised metered dose inhaler SD Standard deviations Authors’ contributions JYP conceived the original idea and supervised the work. . MR collected and analysed the data, and took the lead in writing the manuscript as well. IRG provided critical feedback and contributed to the final manuscript. EDP supervised the pharmacological aspect of the study. MG was in charge of overall direction. Funding Open access funding provided by University of Lausanne. Compliance with ethical standards Conflict of interest The authors declare that they have no conflict of interest. Ethical approval This article does not contain any studies with human participants or animals performed by any of the authors. Informed consent Informed consent was obtained from all individual participants included in the study. The original online version of this article was revised: The name of the first author of the above mentioned published article has a double last name. The family name should have been “Ruman-Colombier” instead of “Ruman”. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Change history 3/8/2021 A Correction to this paper has been published: 10.1007/s00431-020-03863-6
ALBUTEROL SULFATE
DrugsGivenReaction
CC BY
33089387
18,487,237
2021-04
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Lactic acidosis'.
Prevalence and risk factors of lactic acidosis in children with acute moderate and severe asthma, a prospective observational study. Lactic acidosis is a common complication of status asthmaticus in adults. However, data is sparse in children. The aim of this study was to describe the prevalence and risk factors for lactic acidosis in children hospitalised for acute moderate or severe asthma. A total of 154 children 2-17 years of age were enrolled in a prospective observational study conducted in a tertiary hospital. All had capillary blood gas assessment 4 h after the first dose of salbutamol in hospital. The primary endpoint was the prevalence of lactic acidosis. Potential contributing factors such as age, sex, BMI, initial degree of asthma severity, type of salbutamol administration (nebuliser or inhaler), steroids, ipratropium bromide, and glucose-containing maintenance fluid represented secondary endpoints. All in all, 87% of patients had hyperlactatemia (lactate concentration > 2.2 mmol/l). Lactic acidosis (lactate concentration > 5 mmol/l and anion gap ≥ 16 mmol/l) was observed in 26%. In multivariate analysis, age more than 6 years (OR = 2.8, 95% CI 1.2-6.6), glycemia above 11 mmol/l (OR = 3.2 95% CI 1.4-7.4), and salbutamol administered by nebuliser (OR = 10, 95% CI 2.7-47) were identified as risk factors for lactic acidosis in children with moderate or severe asthma.Conclusion: Lactic acidosis is a frequent and early complication of acute moderate or severe asthma in children. What is Known: • Lactic acidosis during acute asthma is associated with b2-mimetics administration. • Salbutamol-related lactic acidosis is self-limited but important to recognise, as compensatory hyperventilation of lactic acidosis can be mistaken for respiratory worsening and lead to inappropriate supplemental bronchodilator administration. What is New: • Lactic acidosis is a frequent complication of acute asthma in the paediatric population. • Age older than 6 years, hyperglycaemia, and nebulised salbutamol are risk factors for lactic acidosis during asthma. Introduction Asthma is characterised by chronic airway inflammation and hyperresponsiveness. During an acute exacerbation, inhomogeneous airway narrowing and obstruction lead to hypoxemia. Compensatory hyperventilation mediated by pulmonary mechanoreceptors conducts to respiratory alkalosis, the most frequent acid-base disturbance observed in acute asthma. If not treated properly, progressive respiratory insufficiency may occur with hypercapnia and respiratory acidosis [1]. Lactic acidosis is another blood gas alteration observed during moderate or severe asthma. Multiple mechanisms have been proposed. Some have evoked reduced tissue perfusion or overuse of respiratory muscles under hypoxic conditions [2]. Others have suggested that b2-adrenergic agents like bronchodilators used to treat asthma lead to increased gluconeogenesis, glycogenolysis, glycolysis, and lipolysis, cumulating in lactic acid production [3–6]. Depending on the mechanism of lactate formation, two types of lactic acidosis exist that can be distinguished calculating the lactate to pyruvate ratio (L/P). Type A (L/P ratio < 25/1) is related to impaired oxygenation, and type B (L/P > 25/1) is caused by excessive b2-receptor stimulation [6]. Lactic acidosis is now a well-known complication of status asthmaticus in adults. Case reports and some retrospective and rare prospective studies describe a transient lactic acidosis as a side effect of high doses b2-agonists used in acute asthma treatment. However, data is rare in children [7]. Even if lactic acidosis during asthma is a self-limited condition, it has an impact on assessment and management of respiratory distress. Compensatory hyperventilation of lactic acidosis is often mistaken as a sign of respiratory worsening and leads to inappropriate escalation of bronchodilator therapy, increasing morbidity and mortality [8, 9]. The aim of our study is to describe the prevalence and risk factors contributing to lactic acidosis in children treated with salbutamol for moderate or severe acute asthma. Materials and methods Study design This prospective observational monocentric prevalence study was conducted from May 01, 2017, to April 30, 2019, in a tertiary care children’s hospital. Patients Children and adolescents 2 to 17 years of age hospitalised for acute moderate or severe asthma were eligible for the study. At our institution, patients requiring inhaled b2-agonists minimum every 2 h fill the indications to be admitted, and an initial degree of asthma severity based on PRAM score is part of the clinical evaluation [10]. Exclusion criteria were as follows: parent’s refusal, metabolic disorder, shock, sepsis, renal or hepatic insufficiency, diabetes mellitus, and cancer. Data collection Basic clinical data included age; body mass index (BMI); sex; initial degree of asthma severity; dose and type of salbutamol administration, of oral or intravenous steroids, and of inhaled ipratropium bromide; and type of maintenance intravenous fluid. Hypoxemia (defined as SpO2 < 92%) was rigorously assessed and treated. Salbutamol was administered by using a metered dose inhaler (pMDI) (through a valved holding chamber with a mouthpiece or a mask, when necessary, AeroChamber Plus Flow Vu®, 1push = 100 cmg of salbutamol) or nebuliser (aerosol solution of 5 mg of salbutamol in 5 ml of normal saline solution NaCl 0.9%). Three types of maintenance fluid were used: 91% of G10% and 9% of NaCl 10% mix, 91% of G5% and 9% of NaCl 10% mix, or normal saline solution (NaCl 0.9%). Capillary blood sample was drawn 4 h after the first administration of salbutamol in hospital, or in case of secondary appearance of tachypnoea or worsening of the respiratory status during hospitalisation. Oxygen saturation was measured by pulse oximetry before blood extraction. Blood gas assessment including the measurement of pH, pCO2, HCO3, base excess (BE), and glucose and lactate concentrations was performed on a RAPIDPoint500, Siemens, gasometer. One millilitre of blood was used for analysis. Hyperlactatemia was defined as lactate concentration > 2.2 mmol/l. Lactic acidosis was defined as lactate > 5 mmol/l and anion gap (AG) ≥ 16mmol/l (AG = Na + K-HC03-Cl), non-compensated lactic acidosis as pH < 7.35, and lactate concentration as > 5 mmol/l. Compensated lactic acidosis represented lactate concentration > 5 mmol/l, pH ≥ 7.35, and pCO2 < 35 mmHg. Hyperglycaemia was characterised as glucose > 11 mmol/l. Outcomes The primary outcome was the prevalence of lactic acidosis. Secondary outcomes included other contributing factors like age, sex, BMI, initial degree of severity, salbutamol administered by inhaler or by nebuliser, steroids, ipratropium bromide, and glucose-containing maintenance fluid. Ethical considerations Patients older than 11 years and parents or legal guardians were informed orally and in writing about the research project by one of the doctors working in the paediatric emergency room during admission to the hospital. Adolescents ≥ 14 years and their parents or legal guardians provided informed consent. The study was approved by the institutional ethics committee (Swiss Ethics, protocol number 2016-01320). Statistical analysis Categorical data were described as absolute counts. Percentages and continuous data we described as means and standard deviations (SD). The Mann–Whitney U test was used to compare the means. We firstly realised simple logistic regression and calculated odd ratio for all potential risk factors. All variables potentially able to influence lactic acidosis (p < 0.2) were used as covariates in multiple logistic regression. Pearson’s correlation coefficient was used to measure statistical relationship between the levels of lactates and doses of salbutamol. All statistical analysis was performed with the Epi Info version 7.2.3.1 software (Centres for Disease Control and Prevention). Results Among 627 patients from 2 to 17 years of age hospitalised for moderate or severe asthma, 174 received information about the study. Eleven did not provided informed consent, and 9 did not have blood sample for technical problem. Finally, 154 patients were included in the study (Fig. 1)Fig. 1 Flow chart of study population Among 154 patients, 99 (64%) were aged from 2 to 6 years and 55 (36%) were more than 6. A total of 13% were obese (P > 97‰). Sex ratio was 1.92 male to female. Forty-three percent of episodes were categorised as severe (PRAM > 8) and 57% as moderate (PRAM 4–8). Salbutamol was administered by pMDI with holding chamber alone in 30% of cases (46/154) or by nebuliser (alone or associated with pMDI administration) in 70% (108/154) (Table 1). Mean overall dose of salbutamol administered during the first 4 h after hospital admission was 12 ± 10 mg. The mean dose was similar in older (> 6 years of age) and younger groups (≤ 6 years of age), 12.3 ± 10.5 mg and 11.8 ± 9.8 mg respectively (p = 0.59). Patients with severe asthma received higher doses of salbutamol (mean 19 ± 10.1 mg) compared to patients with moderate asthma (mean 6.67 ± 5.7 mg), p < 0.0005. When delivered by nebulisation (1 nebulisation = 5 mg of salbutamol), the mean dose of salbutamol was 16 ± 9.3 mg versus 2.4 ± 1.2 mg when delivered by inhaler (1 push = 100 cmg), p < 0.0005.Table 1 Characteristics of study population Characteristics n = 154 % Age ≥ 6 years of age 55 36 Female sex 51 33 Severe asthma (PRAM 8–12) 66 43 Moderate asthma (PRAM 4–7) 88 57 PICU admission 0 0 Obesity 20 13 Salbutamol administered by inhaler 46 30 Salbutamol administered by nebuliser 108 70 Intravenous salbutamol 0 0 Aminophylline 0 0 Corticoids 146 95 Ipratropium bromide 23 15 Glucose-containing maintenance fluid 30 19 Hyperlactatemia > 2.2 mmol/l 134 87 Hyperlactatemia > 5 mmol/l 40 26 Non-compensated lactic acidosis (lactate > 5 mmol/l and pH < 7.35 mmHg) 6 4 Compensated lactic acidosis (lactate > 5, pH > 7.35, pCO2 < 35 mmHg) 34 22 Hyperglycaemia 56 36 Almost all patients (95%) received corticosteroids with a mean dose of 1.85 ± 0.75 mg/kg. Corticosteroids were mainly administered orally (88%). Twenty-three of 154 patients (15%) received ipratropium bromide. Nineteen percent were perfused with glucose-containing maintenance fluid (91% of G10% and 9% of NaCl 10% mix or 91% of G5% and 9% of NaCl 10%) (Table 1). All patients had capillary blood gas analysis 4 h after the first dose of salbutamol administration. Secondary appearance of tachypnoea or worsening of the respiratory status motivated a second blood gas analysis in 13 patients. Most of the patients (87%) had mild hyperlactatemia (lactate > 2.2 mmol/l). All of patients with lactate concentration > 5 mmol/l (26%) had lactic acidosis (lactate > 5 mmol/l and augmented anion gap AG ≥ 16 mmol/l). Thirty-four (22%) presented compensated lactic acidosis (lactate > 5 mmol/l, pH ≥ 7.35, and pCO2 < 35 mmHg). Only 6 (4%) had pH < 7.35. None had hypercapnia (pCO2 > 40 mmHg) (Table 1). In univariate analyses, a significant correlation was found between lactic acidosis and female sex (OR = 2, 95% CI 1–4.2), as well as between lactic acidosis and severe asthma (OR = 4, 95% CI 1.9–8.6). When adjusted on potential confusing factors in multivariate analysis, salbutamol administered by nebuliser (aOR = 10, 95% CI 2.7–47), age older than 6 years (aOR = 2.8, 95% CI 1.2–6.6), and hyperglycaemia (aOR = 3.2 95% CI 1.4–7.4) were related to increased risk of lactic acidosis (Table 2).Table 2 Risk factors of lactic acidosis Risk factors of lactic acidosis Number of patients (%) Univariate analysis unadjusted OR [95% CI] Multivariate analysis adjusted OR [95% CI] Lactic acidosis (yes/non) n = 40 n = 114 Female sex 18 (45) 33 (29) 2 [1–4.2] 1.9 [0.8–4.4] Age ≥ 6 years of age 19 (48) 36 (32) 1.9 [1–4.1] 2.8* [1.2–6.6] Severe asthma 27 (68) 39 (34) 4 [1.9–8.6] 2 [0.8–5.2] Obesity 8 (20) 12 (11) 2.1 [0.8–5.7] 2.5 [0.8–8.1] Hyperglycemia (> 11 mmol/l) 24 (60) 32 (28) 3.8 [1.8–8.2] 3.2* [1.4–7.4] Glucose-containing maintenance fluid 8 (20) 22 (19) 1.05 [0.4–2.6] Corticoids 41 (100) 106 (95) Ipratropium bromure 8 (20) 15 (13) 1.7 [0.6–4.3] Salbutamol administered by nebulisation 38 (95) 70 (61) 11.9 [2.7–52] 10* [2.3–47] OR, odds ratio; CI, confidence interval Number of patients was expressed in absolute number and in percentage (%) All variables associated with p < 0.2 were included in the multivariate analysis Significative findings were marked with an asterisk (*p < 0.05) Lactic acidosis was observed even with low doses of salbutamol (< 0.5 mg/kg). No correlation was found between lactate levels and salbutamol doses (Pearson’s r = 0.17 (SE = 0.24)) (Fig. 2).Fig. 2 Relation between lactate levels and doses of salbutamol Discussion Although lactic acidosis is a well-known metabolic disturbance of asthma in adults, data are rare in the paediatric population. To our knowledge, this is the first large prospective study describing prevalence and risk factors for lactic acidosis in children with acute moderate or severe asthma. A total of 87% (134/154) of our patients had increased blood lactate concentration (lactate > 2.2 mmol/l) 4 h after the first salbutamol dose in hospital. Our findings compare to those of Meert who recorded 83% (62/105) of children with mild hyperlactatemia (lactate> 2.2 mmol/l) during status asthmaticus [5]. Lower prevalence (71%) was found in a retrospective study of 75 children, but the time of lactatemia assessment was not specified [11]. In adults, the prevalence of mild hyperlactatemia (lactate > 2 mmol/l) ranges from 59 in 29 patients with severe asthma 4 to 6 h after therapy beginning [12] to 69.2% at an earlier time point for lactate measurement (about 1 h 25 min after albuterol treatment beginning) [13]. In paediatric case reports, lactate concentration in asthma-related lactic acidosis ranged from 5.9 to 9.2 mmol/l [8]. Koul documented lactic acidosis with a peak lactate range (5.2–13 mmol/l) 2–8 h after the beginning of aerosol therapy in 4 children 11 to 17 years of age. In our study, lactatemia varied from 1.4 to 9.66 mmol/l 4 h after salbutamol administration, with 26% of patients presenting respiratory-compensated lactic acidosis (lactate > 5 mmol/l and AG ≥ 16 mmol/l, pCO2 < 35 mmHg), consistent with other observations [14]. In a retrospective study of 75 children with acute asthma, metabolic acidosis (pH < 7.35 and BE < − 7) was found in 21% of patients [11]. Available lactate level was > 5 mmol/l in 22% of children. Likewise, Meert identified 28% of 53 patients with metabolic acidosis (pH < 7.35, PCO2 < 35 mmHg, and BE < − 7 mmol/l) during acute asthma, with lactate assessment from 7.2 to 9.3 mmol/l 8 to 24 h after admission [4]. Lastly, lactic acidosis with or without respiratory compensation was identified in 47 of 105 (45%) children with acute asthma [5]. In critically ill paediatric patients with asthma, acidosis (pH < 7.35) was found in 45% of patients admitted to the intensive care unit (ICU). Only one had metabolic acidosis with hyperlactatemia (4.6 mmol/l 6 h after ICU admission). All the others had acidosis from respiratory origin. However, in these patients, the blood gas determination was realised quite early, within 2 h after emergency room admission, and the dose of salbutamol received was not specified. Yousef did not find metabolic acidosis in eight other episodes of severe respiratory failure attributable to asthma and suggested that lactic acidosis during asthma is not underestimated and children may be more resistant than adults to the development of this complication. He implied that metabolic acidosis reported in previous studies could be rather related to ketosis following suboptimal hydration and caloric management [15]. We cannot support this hypothesis because all our patients unable to feed or to drink received intravenous glucose perfusion. Lactic acidosis implies two mechanisms. Type A is associated with impaired oxygen delivery and/or hypotension. Type B implies underlying disease (liver or renal insufficiency, diabetes mellitus, or cancer), drugs (such as b2-agonists), or inborn errors of metabolism [16]. None of our patients had known chronic underlying disease. Many authors thought lactic acidosis during asthma to be type B [4–6]. Exposition to high doses of bronchodilator-type salbutamol induces hyperadrenergic state and leads to increased lactate production. Presence of lactic acidosis in patients receiving b-2 agonist therapy under optimal oxygenation or artificial ventilation supports this hypothesis [17]. On the biological level, types A and B can be distinguished by the L/P ratio (L/P < 25/1 versus L/ P> 25/1, respectively). Meert calculated the L/P ratio and concluded that type B lactic acidosis is the most frequent in asthma [5]. Even if hyperlactatemia has been described as a marker of mortality in critically ill patients [18], type B lactic acidosis is a self-limiting condition, and no fatal case has been described in children. The spontaneous resolution with decreasing doses or discontinuation of bronchodilator therapy is a rule [4, 5]. In our study, we did not perform pyruvate assessment for technical reasons and thus could not ascertain the mechanism of lactic acidosis precisely, but we highly suggest type B because none was hypoxemic at the time of lactate assessment and a favourable evolution was observed for all. In our patients, mean total dose of salbutamol delivered by nebulisation (16 ± 9.3 mg) was almost seven times higher compared to the mean dose delivered by inhalator (2.4 ± 1.2 mg). Higher doses could explain the greater risk of lactic acidosis if salbutamol is administered by nebulisation (RR = 10, 95% CI 2.3–47). According the Cochrane database, other side effects of salbutamol such as increased pulse rate were lower for pMDI in children (mean difference − 5% baseline, 95% CI − 8 to − 2%), as was the risk of developing tremor (RR = 0.64; 95% CI 0.44 to 0.95) [19]. On the other hand, in the paediatric population, only 1–10% nebulised salbutamol reaches the inferior respiratory tract [20–23]. Previous study by Wildhaber has shown equivalent percentages of total lung deposition of radiolabeled salbutamol aerosolised by using either a nebuliser or a pMDI with holding chamber (9.6% and 11% for inhaled and nebulised respectively in children > 4 years of age and 5.4% for both in children < 4 years of age) [22]. In a more recent study, it was shown that the amount of drug delivered from pMDI was higher, ranging from 18.1 to 22.5% in young children (3–5 years of age) [22, 23] and from 35.4 to 54.9% in older children (5–17 years of age) [24]. The authors concluded that most children from 5 years of age could obtain lung deposition of more than 30% using a tidal breathing technique with a pMDI. All our patients receiving salbutamol by inhaler via pMDI used this inhalation technique. We did not find a correlation between lactate levels and doses of salbutamol. The statistical power of a dose correlation would be probably reduced by a large proportion of children with mild hyperlactatemia (2.2–5 mmol/l). Intravenous, oral, and inhaled salbutamol raise glycogenolysis resulting in hyperglycaemia. Concurrent use of corticosteroids may exacerbate blood glucose level [6]. In our study, almost all patients received concomitant steroids (95%), and 36% of them had hyperglycaemia (glucose > 11 mmol/l). b2-agonist drugs have two main actions. Firstly, b2-adrenergic receptor stimulation increases glycogenolysis, neoglucogenesis, and glycolysis leading to transformation of glucose to glucose 6-phosphate and then to pyruvate. Secondly, b2-agonists enhance lipolysis. Free acids inhibit pyruvate dehydrogenase, an enzyme which normally allows pyruvate to enter the Krebs cycle. In this way, pyruvate to lactate formation is promoted [25]. In our study, we show that hyperglycaemia raises the risk of lactic acidosis during asthma (aOR = 3.2 95% CI 1.4–7.4). On the intracellular level, the rise in glucose blood level via bronchodilator-mediated glycolysis provides more substrate for lactate production. In patients with severe asthma in ICU, serum glucose was measured. Even if 88% of them had hyperglycaemia (> 6.8 mmol/l), the relationship between hyperglycaemia and lactate concentration could not be proved [4]. Our study showed that children aged more than 6 have almost three times more risk to develop lactic acidosis during asthma (aOR = 2.8, 95% CI 1.2–6.6), despite the fact that the mean dose of salbutamol was the same in both age groups (mean of 11.8 mg, SD 9.8 for < 6 years versus 12.3 mg, SD 10.5 for > 6 years). It could be related to the fact that younger children dispose less glucose resources and in consequence less substrate for lactate production. We identified other parameters like female sex, severe asthma, and obesity as independent risk factors for lactic acidosis. Even if they could not be confirmed in multivariate analysis, they need to be put forward as our study is the first one to try to identify potential risk factors of lactic acidosis in children with asthma. Our study has some limits. Firstly, only 25% of potential patients with moderate or severe asthma seen in the emergency room were included, which is a major selection bias. Secondly, we performed only capillary lactate assessment. However, prior research suggests that capillary lactate value accurately reflects arterial lactate [26]. Moreover, this technique is quicker and easier to perform, especially in children. Another limitation is the lack of initial lactate level. It would be interesting to compare the lactate level upon arrival and 4 h later. Finally, the relationship between steroid therapy and lactic acidosis could not be investigated because almost all patient received corticosteroids. Conclusion Lactic acidosis is a frequent and early (H4) complication of asthma observed in children treated with high doses of bronchodilators. Salbutamol administered by nebuliser, age more than 6 years, and hyperglycaemia were identified as risk factors of lactic acidosis during asthma. Even if self-limited, this condition is important to recognise to avoid unnecessary and harmful therapeutic intensification. Abbreviations AG Anion gap aOR Adjusted odds ratio BE Base excess BMI Body mass index CI Confidence interval L/P Lactate to pyruvate ratio OR Odds ratio pMDI Pressurised metered dose inhaler SD Standard deviations Authors’ contributions JYP conceived the original idea and supervised the work. . MR collected and analysed the data, and took the lead in writing the manuscript as well. IRG provided critical feedback and contributed to the final manuscript. EDP supervised the pharmacological aspect of the study. MG was in charge of overall direction. Funding Open access funding provided by University of Lausanne. Compliance with ethical standards Conflict of interest The authors declare that they have no conflict of interest. Ethical approval This article does not contain any studies with human participants or animals performed by any of the authors. Informed consent Informed consent was obtained from all individual participants included in the study. The original online version of this article was revised: The name of the first author of the above mentioned published article has a double last name. The family name should have been “Ruman-Colombier” instead of “Ruman”. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Change history 3/8/2021 A Correction to this paper has been published: 10.1007/s00431-020-03863-6
ALBUTEROL
DrugsGivenReaction
CC BY
33089387
18,517,670
2021-04
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Metabolic acidosis'.
Prevalence and risk factors of lactic acidosis in children with acute moderate and severe asthma, a prospective observational study. Lactic acidosis is a common complication of status asthmaticus in adults. However, data is sparse in children. The aim of this study was to describe the prevalence and risk factors for lactic acidosis in children hospitalised for acute moderate or severe asthma. A total of 154 children 2-17 years of age were enrolled in a prospective observational study conducted in a tertiary hospital. All had capillary blood gas assessment 4 h after the first dose of salbutamol in hospital. The primary endpoint was the prevalence of lactic acidosis. Potential contributing factors such as age, sex, BMI, initial degree of asthma severity, type of salbutamol administration (nebuliser or inhaler), steroids, ipratropium bromide, and glucose-containing maintenance fluid represented secondary endpoints. All in all, 87% of patients had hyperlactatemia (lactate concentration > 2.2 mmol/l). Lactic acidosis (lactate concentration > 5 mmol/l and anion gap ≥ 16 mmol/l) was observed in 26%. In multivariate analysis, age more than 6 years (OR = 2.8, 95% CI 1.2-6.6), glycemia above 11 mmol/l (OR = 3.2 95% CI 1.4-7.4), and salbutamol administered by nebuliser (OR = 10, 95% CI 2.7-47) were identified as risk factors for lactic acidosis in children with moderate or severe asthma.Conclusion: Lactic acidosis is a frequent and early complication of acute moderate or severe asthma in children. What is Known: • Lactic acidosis during acute asthma is associated with b2-mimetics administration. • Salbutamol-related lactic acidosis is self-limited but important to recognise, as compensatory hyperventilation of lactic acidosis can be mistaken for respiratory worsening and lead to inappropriate supplemental bronchodilator administration. What is New: • Lactic acidosis is a frequent complication of acute asthma in the paediatric population. • Age older than 6 years, hyperglycaemia, and nebulised salbutamol are risk factors for lactic acidosis during asthma. Introduction Asthma is characterised by chronic airway inflammation and hyperresponsiveness. During an acute exacerbation, inhomogeneous airway narrowing and obstruction lead to hypoxemia. Compensatory hyperventilation mediated by pulmonary mechanoreceptors conducts to respiratory alkalosis, the most frequent acid-base disturbance observed in acute asthma. If not treated properly, progressive respiratory insufficiency may occur with hypercapnia and respiratory acidosis [1]. Lactic acidosis is another blood gas alteration observed during moderate or severe asthma. Multiple mechanisms have been proposed. Some have evoked reduced tissue perfusion or overuse of respiratory muscles under hypoxic conditions [2]. Others have suggested that b2-adrenergic agents like bronchodilators used to treat asthma lead to increased gluconeogenesis, glycogenolysis, glycolysis, and lipolysis, cumulating in lactic acid production [3–6]. Depending on the mechanism of lactate formation, two types of lactic acidosis exist that can be distinguished calculating the lactate to pyruvate ratio (L/P). Type A (L/P ratio < 25/1) is related to impaired oxygenation, and type B (L/P > 25/1) is caused by excessive b2-receptor stimulation [6]. Lactic acidosis is now a well-known complication of status asthmaticus in adults. Case reports and some retrospective and rare prospective studies describe a transient lactic acidosis as a side effect of high doses b2-agonists used in acute asthma treatment. However, data is rare in children [7]. Even if lactic acidosis during asthma is a self-limited condition, it has an impact on assessment and management of respiratory distress. Compensatory hyperventilation of lactic acidosis is often mistaken as a sign of respiratory worsening and leads to inappropriate escalation of bronchodilator therapy, increasing morbidity and mortality [8, 9]. The aim of our study is to describe the prevalence and risk factors contributing to lactic acidosis in children treated with salbutamol for moderate or severe acute asthma. Materials and methods Study design This prospective observational monocentric prevalence study was conducted from May 01, 2017, to April 30, 2019, in a tertiary care children’s hospital. Patients Children and adolescents 2 to 17 years of age hospitalised for acute moderate or severe asthma were eligible for the study. At our institution, patients requiring inhaled b2-agonists minimum every 2 h fill the indications to be admitted, and an initial degree of asthma severity based on PRAM score is part of the clinical evaluation [10]. Exclusion criteria were as follows: parent’s refusal, metabolic disorder, shock, sepsis, renal or hepatic insufficiency, diabetes mellitus, and cancer. Data collection Basic clinical data included age; body mass index (BMI); sex; initial degree of asthma severity; dose and type of salbutamol administration, of oral or intravenous steroids, and of inhaled ipratropium bromide; and type of maintenance intravenous fluid. Hypoxemia (defined as SpO2 < 92%) was rigorously assessed and treated. Salbutamol was administered by using a metered dose inhaler (pMDI) (through a valved holding chamber with a mouthpiece or a mask, when necessary, AeroChamber Plus Flow Vu®, 1push = 100 cmg of salbutamol) or nebuliser (aerosol solution of 5 mg of salbutamol in 5 ml of normal saline solution NaCl 0.9%). Three types of maintenance fluid were used: 91% of G10% and 9% of NaCl 10% mix, 91% of G5% and 9% of NaCl 10% mix, or normal saline solution (NaCl 0.9%). Capillary blood sample was drawn 4 h after the first administration of salbutamol in hospital, or in case of secondary appearance of tachypnoea or worsening of the respiratory status during hospitalisation. Oxygen saturation was measured by pulse oximetry before blood extraction. Blood gas assessment including the measurement of pH, pCO2, HCO3, base excess (BE), and glucose and lactate concentrations was performed on a RAPIDPoint500, Siemens, gasometer. One millilitre of blood was used for analysis. Hyperlactatemia was defined as lactate concentration > 2.2 mmol/l. Lactic acidosis was defined as lactate > 5 mmol/l and anion gap (AG) ≥ 16mmol/l (AG = Na + K-HC03-Cl), non-compensated lactic acidosis as pH < 7.35, and lactate concentration as > 5 mmol/l. Compensated lactic acidosis represented lactate concentration > 5 mmol/l, pH ≥ 7.35, and pCO2 < 35 mmHg. Hyperglycaemia was characterised as glucose > 11 mmol/l. Outcomes The primary outcome was the prevalence of lactic acidosis. Secondary outcomes included other contributing factors like age, sex, BMI, initial degree of severity, salbutamol administered by inhaler or by nebuliser, steroids, ipratropium bromide, and glucose-containing maintenance fluid. Ethical considerations Patients older than 11 years and parents or legal guardians were informed orally and in writing about the research project by one of the doctors working in the paediatric emergency room during admission to the hospital. Adolescents ≥ 14 years and their parents or legal guardians provided informed consent. The study was approved by the institutional ethics committee (Swiss Ethics, protocol number 2016-01320). Statistical analysis Categorical data were described as absolute counts. Percentages and continuous data we described as means and standard deviations (SD). The Mann–Whitney U test was used to compare the means. We firstly realised simple logistic regression and calculated odd ratio for all potential risk factors. All variables potentially able to influence lactic acidosis (p < 0.2) were used as covariates in multiple logistic regression. Pearson’s correlation coefficient was used to measure statistical relationship between the levels of lactates and doses of salbutamol. All statistical analysis was performed with the Epi Info version 7.2.3.1 software (Centres for Disease Control and Prevention). Results Among 627 patients from 2 to 17 years of age hospitalised for moderate or severe asthma, 174 received information about the study. Eleven did not provided informed consent, and 9 did not have blood sample for technical problem. Finally, 154 patients were included in the study (Fig. 1)Fig. 1 Flow chart of study population Among 154 patients, 99 (64%) were aged from 2 to 6 years and 55 (36%) were more than 6. A total of 13% were obese (P > 97‰). Sex ratio was 1.92 male to female. Forty-three percent of episodes were categorised as severe (PRAM > 8) and 57% as moderate (PRAM 4–8). Salbutamol was administered by pMDI with holding chamber alone in 30% of cases (46/154) or by nebuliser (alone or associated with pMDI administration) in 70% (108/154) (Table 1). Mean overall dose of salbutamol administered during the first 4 h after hospital admission was 12 ± 10 mg. The mean dose was similar in older (> 6 years of age) and younger groups (≤ 6 years of age), 12.3 ± 10.5 mg and 11.8 ± 9.8 mg respectively (p = 0.59). Patients with severe asthma received higher doses of salbutamol (mean 19 ± 10.1 mg) compared to patients with moderate asthma (mean 6.67 ± 5.7 mg), p < 0.0005. When delivered by nebulisation (1 nebulisation = 5 mg of salbutamol), the mean dose of salbutamol was 16 ± 9.3 mg versus 2.4 ± 1.2 mg when delivered by inhaler (1 push = 100 cmg), p < 0.0005.Table 1 Characteristics of study population Characteristics n = 154 % Age ≥ 6 years of age 55 36 Female sex 51 33 Severe asthma (PRAM 8–12) 66 43 Moderate asthma (PRAM 4–7) 88 57 PICU admission 0 0 Obesity 20 13 Salbutamol administered by inhaler 46 30 Salbutamol administered by nebuliser 108 70 Intravenous salbutamol 0 0 Aminophylline 0 0 Corticoids 146 95 Ipratropium bromide 23 15 Glucose-containing maintenance fluid 30 19 Hyperlactatemia > 2.2 mmol/l 134 87 Hyperlactatemia > 5 mmol/l 40 26 Non-compensated lactic acidosis (lactate > 5 mmol/l and pH < 7.35 mmHg) 6 4 Compensated lactic acidosis (lactate > 5, pH > 7.35, pCO2 < 35 mmHg) 34 22 Hyperglycaemia 56 36 Almost all patients (95%) received corticosteroids with a mean dose of 1.85 ± 0.75 mg/kg. Corticosteroids were mainly administered orally (88%). Twenty-three of 154 patients (15%) received ipratropium bromide. Nineteen percent were perfused with glucose-containing maintenance fluid (91% of G10% and 9% of NaCl 10% mix or 91% of G5% and 9% of NaCl 10%) (Table 1). All patients had capillary blood gas analysis 4 h after the first dose of salbutamol administration. Secondary appearance of tachypnoea or worsening of the respiratory status motivated a second blood gas analysis in 13 patients. Most of the patients (87%) had mild hyperlactatemia (lactate > 2.2 mmol/l). All of patients with lactate concentration > 5 mmol/l (26%) had lactic acidosis (lactate > 5 mmol/l and augmented anion gap AG ≥ 16 mmol/l). Thirty-four (22%) presented compensated lactic acidosis (lactate > 5 mmol/l, pH ≥ 7.35, and pCO2 < 35 mmHg). Only 6 (4%) had pH < 7.35. None had hypercapnia (pCO2 > 40 mmHg) (Table 1). In univariate analyses, a significant correlation was found between lactic acidosis and female sex (OR = 2, 95% CI 1–4.2), as well as between lactic acidosis and severe asthma (OR = 4, 95% CI 1.9–8.6). When adjusted on potential confusing factors in multivariate analysis, salbutamol administered by nebuliser (aOR = 10, 95% CI 2.7–47), age older than 6 years (aOR = 2.8, 95% CI 1.2–6.6), and hyperglycaemia (aOR = 3.2 95% CI 1.4–7.4) were related to increased risk of lactic acidosis (Table 2).Table 2 Risk factors of lactic acidosis Risk factors of lactic acidosis Number of patients (%) Univariate analysis unadjusted OR [95% CI] Multivariate analysis adjusted OR [95% CI] Lactic acidosis (yes/non) n = 40 n = 114 Female sex 18 (45) 33 (29) 2 [1–4.2] 1.9 [0.8–4.4] Age ≥ 6 years of age 19 (48) 36 (32) 1.9 [1–4.1] 2.8* [1.2–6.6] Severe asthma 27 (68) 39 (34) 4 [1.9–8.6] 2 [0.8–5.2] Obesity 8 (20) 12 (11) 2.1 [0.8–5.7] 2.5 [0.8–8.1] Hyperglycemia (> 11 mmol/l) 24 (60) 32 (28) 3.8 [1.8–8.2] 3.2* [1.4–7.4] Glucose-containing maintenance fluid 8 (20) 22 (19) 1.05 [0.4–2.6] Corticoids 41 (100) 106 (95) Ipratropium bromure 8 (20) 15 (13) 1.7 [0.6–4.3] Salbutamol administered by nebulisation 38 (95) 70 (61) 11.9 [2.7–52] 10* [2.3–47] OR, odds ratio; CI, confidence interval Number of patients was expressed in absolute number and in percentage (%) All variables associated with p < 0.2 were included in the multivariate analysis Significative findings were marked with an asterisk (*p < 0.05) Lactic acidosis was observed even with low doses of salbutamol (< 0.5 mg/kg). No correlation was found between lactate levels and salbutamol doses (Pearson’s r = 0.17 (SE = 0.24)) (Fig. 2).Fig. 2 Relation between lactate levels and doses of salbutamol Discussion Although lactic acidosis is a well-known metabolic disturbance of asthma in adults, data are rare in the paediatric population. To our knowledge, this is the first large prospective study describing prevalence and risk factors for lactic acidosis in children with acute moderate or severe asthma. A total of 87% (134/154) of our patients had increased blood lactate concentration (lactate > 2.2 mmol/l) 4 h after the first salbutamol dose in hospital. Our findings compare to those of Meert who recorded 83% (62/105) of children with mild hyperlactatemia (lactate> 2.2 mmol/l) during status asthmaticus [5]. Lower prevalence (71%) was found in a retrospective study of 75 children, but the time of lactatemia assessment was not specified [11]. In adults, the prevalence of mild hyperlactatemia (lactate > 2 mmol/l) ranges from 59 in 29 patients with severe asthma 4 to 6 h after therapy beginning [12] to 69.2% at an earlier time point for lactate measurement (about 1 h 25 min after albuterol treatment beginning) [13]. In paediatric case reports, lactate concentration in asthma-related lactic acidosis ranged from 5.9 to 9.2 mmol/l [8]. Koul documented lactic acidosis with a peak lactate range (5.2–13 mmol/l) 2–8 h after the beginning of aerosol therapy in 4 children 11 to 17 years of age. In our study, lactatemia varied from 1.4 to 9.66 mmol/l 4 h after salbutamol administration, with 26% of patients presenting respiratory-compensated lactic acidosis (lactate > 5 mmol/l and AG ≥ 16 mmol/l, pCO2 < 35 mmHg), consistent with other observations [14]. In a retrospective study of 75 children with acute asthma, metabolic acidosis (pH < 7.35 and BE < − 7) was found in 21% of patients [11]. Available lactate level was > 5 mmol/l in 22% of children. Likewise, Meert identified 28% of 53 patients with metabolic acidosis (pH < 7.35, PCO2 < 35 mmHg, and BE < − 7 mmol/l) during acute asthma, with lactate assessment from 7.2 to 9.3 mmol/l 8 to 24 h after admission [4]. Lastly, lactic acidosis with or without respiratory compensation was identified in 47 of 105 (45%) children with acute asthma [5]. In critically ill paediatric patients with asthma, acidosis (pH < 7.35) was found in 45% of patients admitted to the intensive care unit (ICU). Only one had metabolic acidosis with hyperlactatemia (4.6 mmol/l 6 h after ICU admission). All the others had acidosis from respiratory origin. However, in these patients, the blood gas determination was realised quite early, within 2 h after emergency room admission, and the dose of salbutamol received was not specified. Yousef did not find metabolic acidosis in eight other episodes of severe respiratory failure attributable to asthma and suggested that lactic acidosis during asthma is not underestimated and children may be more resistant than adults to the development of this complication. He implied that metabolic acidosis reported in previous studies could be rather related to ketosis following suboptimal hydration and caloric management [15]. We cannot support this hypothesis because all our patients unable to feed or to drink received intravenous glucose perfusion. Lactic acidosis implies two mechanisms. Type A is associated with impaired oxygen delivery and/or hypotension. Type B implies underlying disease (liver or renal insufficiency, diabetes mellitus, or cancer), drugs (such as b2-agonists), or inborn errors of metabolism [16]. None of our patients had known chronic underlying disease. Many authors thought lactic acidosis during asthma to be type B [4–6]. Exposition to high doses of bronchodilator-type salbutamol induces hyperadrenergic state and leads to increased lactate production. Presence of lactic acidosis in patients receiving b-2 agonist therapy under optimal oxygenation or artificial ventilation supports this hypothesis [17]. On the biological level, types A and B can be distinguished by the L/P ratio (L/P < 25/1 versus L/ P> 25/1, respectively). Meert calculated the L/P ratio and concluded that type B lactic acidosis is the most frequent in asthma [5]. Even if hyperlactatemia has been described as a marker of mortality in critically ill patients [18], type B lactic acidosis is a self-limiting condition, and no fatal case has been described in children. The spontaneous resolution with decreasing doses or discontinuation of bronchodilator therapy is a rule [4, 5]. In our study, we did not perform pyruvate assessment for technical reasons and thus could not ascertain the mechanism of lactic acidosis precisely, but we highly suggest type B because none was hypoxemic at the time of lactate assessment and a favourable evolution was observed for all. In our patients, mean total dose of salbutamol delivered by nebulisation (16 ± 9.3 mg) was almost seven times higher compared to the mean dose delivered by inhalator (2.4 ± 1.2 mg). Higher doses could explain the greater risk of lactic acidosis if salbutamol is administered by nebulisation (RR = 10, 95% CI 2.3–47). According the Cochrane database, other side effects of salbutamol such as increased pulse rate were lower for pMDI in children (mean difference − 5% baseline, 95% CI − 8 to − 2%), as was the risk of developing tremor (RR = 0.64; 95% CI 0.44 to 0.95) [19]. On the other hand, in the paediatric population, only 1–10% nebulised salbutamol reaches the inferior respiratory tract [20–23]. Previous study by Wildhaber has shown equivalent percentages of total lung deposition of radiolabeled salbutamol aerosolised by using either a nebuliser or a pMDI with holding chamber (9.6% and 11% for inhaled and nebulised respectively in children > 4 years of age and 5.4% for both in children < 4 years of age) [22]. In a more recent study, it was shown that the amount of drug delivered from pMDI was higher, ranging from 18.1 to 22.5% in young children (3–5 years of age) [22, 23] and from 35.4 to 54.9% in older children (5–17 years of age) [24]. The authors concluded that most children from 5 years of age could obtain lung deposition of more than 30% using a tidal breathing technique with a pMDI. All our patients receiving salbutamol by inhaler via pMDI used this inhalation technique. We did not find a correlation between lactate levels and doses of salbutamol. The statistical power of a dose correlation would be probably reduced by a large proportion of children with mild hyperlactatemia (2.2–5 mmol/l). Intravenous, oral, and inhaled salbutamol raise glycogenolysis resulting in hyperglycaemia. Concurrent use of corticosteroids may exacerbate blood glucose level [6]. In our study, almost all patients received concomitant steroids (95%), and 36% of them had hyperglycaemia (glucose > 11 mmol/l). b2-agonist drugs have two main actions. Firstly, b2-adrenergic receptor stimulation increases glycogenolysis, neoglucogenesis, and glycolysis leading to transformation of glucose to glucose 6-phosphate and then to pyruvate. Secondly, b2-agonists enhance lipolysis. Free acids inhibit pyruvate dehydrogenase, an enzyme which normally allows pyruvate to enter the Krebs cycle. In this way, pyruvate to lactate formation is promoted [25]. In our study, we show that hyperglycaemia raises the risk of lactic acidosis during asthma (aOR = 3.2 95% CI 1.4–7.4). On the intracellular level, the rise in glucose blood level via bronchodilator-mediated glycolysis provides more substrate for lactate production. In patients with severe asthma in ICU, serum glucose was measured. Even if 88% of them had hyperglycaemia (> 6.8 mmol/l), the relationship between hyperglycaemia and lactate concentration could not be proved [4]. Our study showed that children aged more than 6 have almost three times more risk to develop lactic acidosis during asthma (aOR = 2.8, 95% CI 1.2–6.6), despite the fact that the mean dose of salbutamol was the same in both age groups (mean of 11.8 mg, SD 9.8 for < 6 years versus 12.3 mg, SD 10.5 for > 6 years). It could be related to the fact that younger children dispose less glucose resources and in consequence less substrate for lactate production. We identified other parameters like female sex, severe asthma, and obesity as independent risk factors for lactic acidosis. Even if they could not be confirmed in multivariate analysis, they need to be put forward as our study is the first one to try to identify potential risk factors of lactic acidosis in children with asthma. Our study has some limits. Firstly, only 25% of potential patients with moderate or severe asthma seen in the emergency room were included, which is a major selection bias. Secondly, we performed only capillary lactate assessment. However, prior research suggests that capillary lactate value accurately reflects arterial lactate [26]. Moreover, this technique is quicker and easier to perform, especially in children. Another limitation is the lack of initial lactate level. It would be interesting to compare the lactate level upon arrival and 4 h later. Finally, the relationship between steroid therapy and lactic acidosis could not be investigated because almost all patient received corticosteroids. Conclusion Lactic acidosis is a frequent and early (H4) complication of asthma observed in children treated with high doses of bronchodilators. Salbutamol administered by nebuliser, age more than 6 years, and hyperglycaemia were identified as risk factors of lactic acidosis during asthma. Even if self-limited, this condition is important to recognise to avoid unnecessary and harmful therapeutic intensification. Abbreviations AG Anion gap aOR Adjusted odds ratio BE Base excess BMI Body mass index CI Confidence interval L/P Lactate to pyruvate ratio OR Odds ratio pMDI Pressurised metered dose inhaler SD Standard deviations Authors’ contributions JYP conceived the original idea and supervised the work. . MR collected and analysed the data, and took the lead in writing the manuscript as well. IRG provided critical feedback and contributed to the final manuscript. EDP supervised the pharmacological aspect of the study. MG was in charge of overall direction. Funding Open access funding provided by University of Lausanne. Compliance with ethical standards Conflict of interest The authors declare that they have no conflict of interest. Ethical approval This article does not contain any studies with human participants or animals performed by any of the authors. Informed consent Informed consent was obtained from all individual participants included in the study. The original online version of this article was revised: The name of the first author of the above mentioned published article has a double last name. The family name should have been “Ruman-Colombier” instead of “Ruman”. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Change history 3/8/2021 A Correction to this paper has been published: 10.1007/s00431-020-03863-6
ALBUTEROL SULFATE
DrugsGivenReaction
CC BY
33089387
18,487,237
2021-04
What was the administration route of drug 'OXYCODONE'?
Considering additive effects of polypharmacy : Analysis of adverse events in geriatric patients in long-term care facilities. BACKGROUND Potential additive effects of polypharmacy are rarely considered in adverse events of geriatric patients living in long-term care facilities. Our aim, therefore, was to identify adverse events in this setting and to assess plausible concomitant drug causes. METHODS A cross-sectional observational study was performed in three facilities as follows: (i) adverse event identification: we structurally identified adverse events using nurses' interviews and chart review. (ii) Analysis of the concomitantly administered drugs per patient was performed in two ways: (ii.a) a review of summary of product characteristics for listed adverse drug reactions to identify possible causing drugs and (ii.b) a causality assessment according to Naranjo algorithm. RESULTS (i) We found 424 adverse events with a median of 4 per patient (range 1-14) in 103 of the 104 enrolled patients (99%). (ii.a) We identified a median of 3 drugs (range 0-11) with actually occurring adverse events listed as an adverse drug reaction in the summary of product characteristics. (ii.b) Causality was classified in 198 (46.9%) of adverse events as "doubtful," in 218 (51.2%) as "possible," in 7 (1.7%) as "probable," and in 1 (0.2%) adverse event as a "definitive" cause of the administered drugs. In 340 (80.2%) of all identified adverse events several drugs simultaneously reached the highest respective Naranjo score. CONCLUSIONS Patients in long-term facilities frequently suffer from many adverse events. Concomitantly administered drugs have to be frequently considered as plausible causes for adverse events. These additive effects of drugs should be more focused in patient care and research. Introduction Geriatric patients in long-term care (LTC) facilities are multimorbid and, therefore, suffer from many (non)specific symptoms and geriatric syndromes [1]. Disease-related symptoms should be distinguished from adverse drug reactions (ADR) that result from drug therapy [2]. The latter can lead to hospital admissions and have a considerable impact on morbidity and mortality with high costs in the health care system [3–5]. Polypharmacy makes a significant contribution to the clinical consequences deriving from ADRs in geriatric patients [6]. For this reason, a structured analysis of adverse events (AE) and drug-related causes in these patients is of high interest for routine care. Distinguishing whether an observed AE was caused by a disease (i.e. symptom) or by a drug (ADR) poses a challenge for healthcare professionals [7, 8]. The correct attribution is required for appropriate treatment strategies but can result only from structured detection, analysis and classification. Geriatric patients are frequently cognitively impaired or suffer from speech or hearing disorders. Hence, information provided by the patients is often insufficient. In LTC facilities, therefore, chart documentation and nurses’ interviews are the most valuable sources for AE detection [9, 10]. So far, no specific method exists to analyze and classify AE in LTC facility residents with polypharmacy. The Naranjo algorithm has previously been used for causality assessment in this collective [11, 12]. It allows a detailed assessment of every detected AE and every single administered drug. This algorithm provides further information on drug-related causes when combined with established methods for patient safety, such as drug-drug interactions and potentially inappropriate medications [13]. Causality scores like the Naranjo score, however, do not consider simultaneously contributing drugs. For some ADRs, it has been shown that the number of specific drugs causes their clinical manifestations. For example, patients are exposed to an increased risk of falling when they take two or more drugs which increase the risk of falling [14]. Concerning anticholinergic ADRs, it is common to calculate an anticholinergic burden to quantify the risk for an adverse outcome [15]. Little is known, however, about additive drug effects in other events. Therefore, data about potential additive effects in this vulnerable patient collective are of great interest for routine care. The aim of this study was to identify AEs occurring in LTC facility patients and to assess plausible concomitant drug causes. Patients, material and methods Definitions We defined an AE as an outcome that occurs while a patient is taking a drug, but is not or not necessarily attributable to it and an ADR as an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product [16]. We used the term drug not only for the effective substance but for the whole product prescribed in the medication chart of the patient. A drug therefore could contain more than one active substance. We considered all drugs administered to the patient during the acquisition period. Continuous and on-demand medications were assessed separately because the temporal relationship between AE and administration of the drug could be different in that case. Participants and setting We conducted a cross-sectional observation study in three LTC facilities in Germany. After written informed consent of the residents or their legal representative and the responsible general practitioner, residents in the participating LTC facilities were enrolled in the study. We included residents of facilities with different ownerships (welfare, municipal or private associations) to approach a representative sample of 100 residents. Inclusion criteria were: informed consent, age ≥65 years, long-term/chronic medicines ≥3 and multimorbidity with ≥3 comorbidities at the time of recruitment, more than 8 weeks stay in the LTC facility, and a life expectancy of more than 6 months according to nurses’ present information. The study was conducted over a time period of 10 months. Study design and data collection We conducted a structured analysis of AEs. (i) AE identification We used two complement sources of information for our structured data collection: Firstly, an interview about individual AEs with nurses involved in daily care and, secondly, a review of residents’ records (electronic and chart documentation, laboratory values) for documented events and their temporal occurrence. To ensure standardized identification of AEs, a checklist of events was applied to both methods. The listed events comprised the most relevant AEs or ADRs for geriatric patients and LTC residents based on the literature [17–19]: blackened stool, bleeding/hematoma, confusion/disorientation, constipation, depression/anxiety, diarrhea, dizziness/vertigo, dry mouth, ear disorders, eye disorders, falls, hallucination, hyperglycemia/hypoglycemia, hyperhidrosis, hyperkalemia/hypokalemia, hypernatremia/hyponatremia, nausea, pain, restlessness, skin disorders/pruritus, insomnia, urinary incontinence, vomiting (in alphabetical order). Additional relevant reported or documented events were collected as well. We considered reported and documented symptoms during a time period of the prior 30 days (considered as 1 resident month) for new and continuous symptoms. Data collection was performed at two measurement points per patient at intervals of 6–8 weeks by two clinical pharmacists. All detected AEs and the corresponding system organ class were classified based on the common terminology criteria for adverse events (CTCAE) [20]. (iia) Review of summary of product characteristics We systematically collected data from the medical charts (continuous drugs, on demand drugs and their frequency of use, date of first administration). We checked all summaries of product characteristics (SmPCs) of the actually administered drugs for listed ADRs. A drug would be considered as “potentially causing” if the listed ADR in the SmPC represented a synonym for the detected AE or possibly caused it (e.g. dizziness in cases of falls). For the analysis of additive effects, we counted the number of potentially causing drugs. Prescribed drugs were characterized by their code in the anatomical therapeutic chemical classification system (ATC code). (iib) Causality assessment according to the Naranjo algorithm We used the Naranjo algorithm for causality assessment. All further relevant information, such as the duration of the AE, underlying diseases, clinical consequences (e.g. from hospital report), laboratory values, and patient-specific conditions were collected and used to determine the Naranjo score. The most likely associated drugs were the ones that reached the highest Naranjo score concerning the single analyzed AE. Naranjo distinguishes between definitive with a total score ≥9, probable with 5 < total score < 8, possible with 1 < total score < 4 and doubtful with a total score ≤ 0 [21, 22]. Inconclusive evaluations in all steps (i, ii.a, and ii.b) were discussed and finalized by mutual agreement in an expert panel. This panel consisted of four experienced clinical pharmacists. Statistical analysis To ensure comparable patient parameters between the three LTC facilities independent of the allocation to a single facility, main patient parameters were statistically analyzed. For this purpose, a Kruskal Wallis test with pairwise comparison was performed. Analyzed parameters were age, gender, number of diagnoses and number of continuous and on demand drugs, as well as the number of AEs in the patients and the maximum Naranjo score per patient. The data analysis was performed using IBM SPSS Statistics Version 25.0 (IBM Corporation, Armonk, NY, USA) and Microsoft Office Excel 2013 (Microsoft Corporation, Redmond, WA, USA). P-values ≤ 0.05 were considered as statistically significant. Results Patient characteristics In the participating parts of the LTC facilities, 182 patients were potentially available for the study and 154 met the inclusion criteria. From these, 104 patients or their legal guardian gave their informed consent as well as their responsible physician and were enrolled in the study. Patients were mostly female (72.1%) and in median 86 (range: 66–101) years old (Table 1). Patients did not differ between the three LTC facilities according to the following parameters: age (p = 0.311), gender (p = 0.684), number of diagnoses (p = 0.070) and number of continuous (p = 0.629) and on demand drugs (p = 0.911).Table 1 Characteristics of patients included in the study with frequency of documented diagnoses, main ATC classes and main active substances Characteristics Value Patients, total, n 104 Patients in facility of welfare ownership, n (%) 34 (32.7%) Patients in facility of municipal ownership, n (%) 30 (28.8%) Patients in facility of ownership by private association, n (%) 40 (38.5%) Female, n (%) 75 (72.1%) Length of residence (months), median (Q25/Q75; min–max) 31 (12/63; 1–414) Age (years), median (Q25/Q75; min–max) 86 (78/90; 66–101) Documented diagnoses, median (Q25/Q75; min–max) 15 (10/21; 3–35) No. of continuous drugs, median (Q25/Q75; min–max) 8 (6/10; 2–18) No. of on demand medication, median (Q25/Q75; min–max) 2 (1/3; 1–6) Documented diagnosisa Hypertension, n (%) 82 (78.8%) Dementia, n (%) 69 (66.3%) Diabetes, n (%) 41 (39.4%) Heart failure, n (%) 32 (30.8%) Atrial fibrillation, n (%) 32 (30.8%) Renal failure, n (%) 24 (23.1%) Osteoporosis, n (%) 19 (18.3%) Stroke, n (%) 17 (16.3%) Main ATC classesb C (cardiovascular system), n (%) 236 (28.7%) N (nervous system), n (%) 216 (26.3%) A (alimentary tract and metabolism), n (%) 164 (20.0%) B (blood and blood-forming organs), n (%) 68 (8.3%) H (systemic hormonal preparations, excluding sex hormones and insulins), n (%) 27 (3.3%) Main active substancesb Torasemide, n (%) 47 (5.7%) Pantoprazole, n (%) 40 (4.9%) Ramipril, n (%) 35 (4.3%) Acetylsalicylic acid, n (%) 33 (4.0%) Metoprolol, n (%) 23 (2.8%) ATC anatomical therapeutic chemical/defined daily dose classification, Q25/Q75 first and third quartile aOrder is based on the most relevant diagnoses found in literature data to geriatric patients bAccording to the documented continuous drugs (i) AE identification From a total of 104 patients, at least 1 AE was identified in 103 (99.0%). We identified 424 AEs, with a detected median of 4 (Q25/Q75: 2/5, range 1–14) AEs per patient, which equals 2.05 AEs per resident month. The identified AEs and the number of affected patients are shown in Table 2. The system organ classes renal and urinary disorder (87 patients), gastrointestinal disorder (43 patients), skin and subcutaneous tissue disorders (37 patients) were most common in our patient collective. Altogether, 72 different AE categories were detected, 185 AEs were identified in the patient records and 195 AEs by the nurses’ interviews, with 44 AEs in concordance of both methods. We found a significant difference in the detected number of AEs between the observed LTC facilities (p = 0.020). Following the pairwise comparison, we only found differences between the municipal LTC facility with 3 (Q25/Q75: 2/4) AEs and the private LTC facility with a median of 4 (Q25/Q75: 3/6.25) AEs (p = 0.022).Table 2 Identified adverse evnts (n = 424) according to CTCAE and affected patients (n = 104) System organ class Number of identified AEs, n (%) Affected patients, n (%) AE with number of affected patientsa (n) Renal and urinary disorders 88 (20.8) 87 (83.7) Urinary incontinence (87), urinary tract pain (1) Gastrointestinal disorders 55 (13.0) 43 (41.3) Constipation (22), vomiting (16), diarrhea (10), blackened stools (3), nausea (2), lower gastrointestinal bleeding (1), periodontal disease (1) Psychiatric disorders 55 (13.0) 35 (33.7) Confusion (21), restlessness (12), defensive behavior (8), insomnia (5), depression (4), anxiety (2), hallucinations (1), personality change (1), psychiatric disorders—other specify (1) Skin and subcutaneous tissue disorders 50 (11.8) 37 (35.6) Intertrigo (9), dry skin (7), hyperhidrosis (7), skin ulceration (6), local redness (5), pruritus (4), purpura (4), skin and subcutaneous tissue disorders—other specify (3), skin induration (1), urticaria (2), alopecia (1), angioedema (1) Metabolism and nutritional disorders 41 (9.7) 27 (26.0) Hyperglycemia (26), hypoglycemia (15) Musculoskeletal and connective tissue disorders 40 (9.4) 33 (31.7) Arthralgia (14), pain in extremity (12), back pain (6), arthritis (4), musculoskeletal and connective tissue disorders—other specify (3), general muscle weakness (1) Nervous system disorders 39 (9.2) 31 (29.8) Dizziness (11), somnolence (10), headache (4), syncope (3), ataxia (2), cognitive disturbance (2), paresthesia (2), depressed level of consciousness (1), lethargy (1), neuralgia (1), seizure (1), spasticity (1) Injury, poisoning and procedural complications 14 (3.3) 14 (13.5) Fall (14) Vascular disorders 11 (2.6) 11 (10.6) Hematoma (10), flushing (1) Infections and infestations 8 (1.9) 7 (6.7) Skin infection (4), vulval infection (2), conjunctivitis infective (1), stoma site infection (1) General disorders and administration site conditions 7 (1.7) 7 (6.7) Edema limbs (3), pain (3), fatigue (1) Respiratory, thoracic and mediastinal disorders 7 (1.7) 6 (5.8) Dyspnea (4), cough (1), epistaxis (1), respiratory, thoracic and mediastinal disorders—other specify (1) Ear and labyrinth disorders 3 (0.7) 3 (2.9) Hearing impaired (2), tinnitus (1) Cardiac disorders 2 (0.5) 2 (1.9) Chest pain—cardiac (1), palpitations (1) Eye disorders 2 (0.5) 1 (1.0) Blurred vision (1), glaucoma (1) Investigations 2 (0.5) 2 (1.9) Weight gain (1), weight loss (1) AE(s) adverse event(s), CTCAE common terminology criteria for adverse events aMultiple categories per patient possible (ii.a) Review of summary of product characteristics To analyze the concomitantly administered drugs, we assessed 3725 combinations of AEs and corresponding drugs. For this analysis five drug/AE pairs had to be excluded because no information from the SmPC was available (moisturizing eye drops, medical device). Considering every identified AE, patients had a median of 3 potentially causing drugs according to the SmPC, with a range from 0 to 11 drugs (Q25/Q75: 2/4; details in Fig. 1). The most frequently (n ≥ 10) detected AEs and the affected system organ classes are shown in Table 3. The ATC classes prescribed most often (C, N, A, B, H) were frequently among the potentially causing drugs for the most common system organ classes (Fig. 2).Fig. 1 Number of detected adverse events versus number of potentially causing drugs according Summary of Product Charactetistics. AE(s) adverse event(s), SmPC summary of products characteristics Table 3 Median number of potentially causing drugs according Summary of Product Charactersitics and corresponding Naranjo Score per patient (n = 104) for the most frequently detected (≥10) adverse events (AEs) and for their corresponding System organ classes (all 424 detected AE included) System organ class and ... most frequent AE Number (n) Median number of potentially causing drugs per patient [range] Median Naranjo score [range] Renal and urinary disorders 88 2 [0–5] 0 [−1–2] ... Urinary incontinence 87 2 [0–5] 0 [−1–2] Gastrointestinal disorders 55 5 [0–11] 2 [0–4] ... Constipation 22 5 [0–10] 0 [0–3] ... Vomiting 16 7.5 [2–10] 2 [0–4] ... Diarrhea 10 4.5 [2–11] 3 [0–3] Psychiatric disorders 55 3 [0–7] 0 [−1–9] ... Confusion 21 3 [1–7] 1 [0–8] ... Restlessness 12 3 [0–4] 0 [−1–3] Metabolism and nutrition disorders 41 3 [0–5] 1 [0–5] ... Hyperglycemia 26 3 [0–4] 1 [0–1] ... Hypoglycemia 15 3 [1–5] 4 [2–5] Musculoskeletal and connective tissue disorders 40 3 [0–8] 2 [−1–3] ... Arthralgia 14 3 [0–6] 1 [−1–3] ... Pain in extremity 12 2 [1–8] 2 [0–3] Nervous system disorders 39 4 [0–10] 1 [−1–7] ... Dizziness 11 5 [1–10] 2 [0–7] ... Somnolence 10 4 [3–7] 3 [0–5] Injury, poisoning and procedural complications 14 6 [3–11] 2 [0–3] ... Fall 14 6 [3–11] 2 [0–3] Vascular disorders 11 1 [0–3] 1 [0–3] ... Hematoma 10 1 [0–3] 0.5 [0–2] AE(s) Adverse Event(s), SmPC Summary of product characteristics Fig. 2 Potentially causing drugs according Summary of Product Characteristics differentiated into the ATC classes for the most frequently detected system organ classes. AE(s) adverse event(s), ATC anatomical therapeutic chemical/defined daily dose classification, CTCAE common criteria for the terminology of adverse events, SmPC summary of products characteristics. ATC classes: A: alimentary tract and metabolism, B: blood and blood forming organs, C: cardiovascular system, H: systemic hormonal preparations, excluding sex hormones and insulins, N: nervous system (ii.b) Causality assessment according to the Naranjo algorithm All 3730 drug/AE pairs were included in the causality assessment. From the 424 identified AEs, 198 (46.9%) were classified as ADR with “doubtful”, 218 (51.2%) “possible”, 7 (1.7%) “probable”, and 1 (0.2%) “definitive” cause (Table 4). We found no significant differences in the maximum Naranjo scores per patient between the three LTC facilities (p = 0.964). On the basis of 424 detected AEs, only 1 drug in 84 AEs (19.8%) and several drugs in 340 AEs (80.2%) reached the highest score (Table 4). According to Naranjo these need to be considered as the most likely causing drug(s).Table 4 Results of the adverse event drug causality assessment according to the Naranjo algorithm Naranjo Score per AE Identified AE [n] Number of affected patientsa, [n] Classification Identified AE per class, n (%) Number of AE with one/several highest Naranjo drug(s) [n] −1 22 17 Doubtful 199 (46.9) 38/161 0 177 92 1 68 48 Possible 217 (51.2) 38/179 2 90 51 3 41 30 4 18 18 5 3 3 Probable 7 (1.7) 7/0 6 2 2 7 1 1 8 1 1 9 1 1 Definitive 1 (0.2) 1/0 AE(s) adverse event(s) aSeveral AEs per patient possible The probable and definitive ADRs were as follows: angioedema (severity grade according to CTCAE 4) induced by enalapril, urticaria (grade 2) induced by amoxicillin and clavulanic acid, hypoglycemia (grade 1) induced by insulin glargine, paresthesia (grade 2) induced by tapentadol and a complex case of occurring hallucinations (grade 3) in combination with confusion (grade 3), dizziness (grade 3) and somnolence (grade 3, in total 4 detected AEs) which were attributable to digitoxin (highest Naranjo score). In this case, the patient also received high doses of oxycodone and duloxetine. It can be seen as a mixed intoxication based on the hospital report. In the algorithm, digitoxin reached a one-point higher score than oxycodone/duloxetine because measurement of the increased blood level was available only for digitoxin. All of the detected AEs and ADRs were managed adequately by the nurses, for example, by informing a physician or arranging a hospital admission for the affected patient. Thus, no further action was required due to this study. Discussion In our study we addressed AEs in geriatric patients living in LTC facilities. We assessed which type of AEs occurred and also investigated potential additive effects of polypharmacy. With nearly all (99%) patients affected by AEs, we demonstrated the relevance of this topic. We found the identified AEs potentially caused by up to 11 different administered drugs. The Naranjo algorithm showed at least possible drug causes in half of these AEs. Thereby, multiple drugs were equally likely involved 80% of the time. Our results point out that AEs should be systematically recorded in routine practice in LTC facilities. In order to prevent ADRs, additive effects need to be considered in any strategies developed. Prevalence of AE and ADR in LTC residents More than half of our identified AEs could be associated with drug use. Our rate of probable and definitive ADRs was similar to other studies in the LTC setting (0.04 vs. up to 0.10 ADRs per observed resident month), although studies should be compared with caution [9, 23]. Nevertheless, the causality assessment leaves us with a high number of possible ADRs. Especially for AEs which were ongoing for a longer period, causality assessment was challenging in the routine setting. Information to evaluate the exact temporal connection between AE and drug use was frequently missing and therefore could have led to lower Naranjo scores. To resolve this problem, a regular and structured routine assessment of AEs and potentially causing drugs might increase the chance to identify ADRs and protect patients from the consequences. Our overall rate of identified AEs was higher than results seen in other studies (2.05 AEs vs 0.03–0.12 per observed resident month) [9, 23]. This indicates that we identified a noticeable amount of the general symptom burden of LTC residents that results from underlying diseases or age-related changes. This is consistent with the fact that incontinence, pain, sleep disorders and psychopathological symptoms are widely found in LTC residents [24]. Therefore, a regular routine AE assessment can support ADR detection as well as structured symptom evaluation. Additive effects of polypharmacy The suspected AE was listed as an ADR in the respective SmPC in a median of 3 and up to 11 administered drugs per patient. In 80% of all identified AEs, various drugs reached the highest Naranjo score simultaneously. This means that they were equally likely to cause the AE. This coincidence can increase the chance of AE occurrence independently from single causality scores. This result also raises the question whether ADRs resulting from additive effects have been underestimated. In cases with “probable” or “definitive” ADRs (Naranjo ≥5), we found results from only one drug with the highest Naranjo score; however, in four of these AEs, the drug with the highest Naranjo (digitoxin) was only part of a mixed intoxication with duloxetine and oxycodone based on the hospital report for the affected patient. In this case, the sole consideration of the causality assessment could mask an additive effect of at least 3 concomitantly given drugs. This shows that additive effects need to be considered in every detected AE independently from the single causality. Besides ADRs from well-known drug classes (e.g. vascular ADRs from drugs affecting blood and blood-forming organs), in a substantial amount of AEs, we found involvement of varying ATC-classes that are less familiar (e.g. nervous system ADRs in drugs affecting the cardiovascular system). This underlines the complexity of geriatric patient treatment and the need for interdisciplinary medication reviews that include an assessment of drug-related problems, such as drug-drug interactions, potentially inappropriate medication, as well as ADRs [25, 26]. In routine care, however, potential additive effects are often not taken into account. In particular, new and unclear symptoms could be misinterpreted as new diseases and sometimes even lead to prescribing cascades [27, 28]. Implications for practice Firstly, our study shows the need for a good data base and a regular routine assessment of AEs that occur in LTC facilities. We found a very low concordance rate of only 10% between AEs detected in nurses’ interviews and those mentioned in the patient record analysis. This demonstrates the potential of information loss in LTC facilities due to heterogeneous and incomplete AE documentation [29]. It also indicates the potential of recall bias in the nurses. The identification of every occurring AE allows a better assessment of simultaneously occurring events. We found in our study, for example, a combination of vomiting and diarrhea that indicated an infection rather than an ADR. Furthermore, the information on patients’ current symptoms contributes to appropriate proposals for medication changes in cases of identified ADRs. Secondly, our results support the development of strategies with improved consideration of the additive effects of polypharmacy. Combining an AE assessment with structured medication reviews improves the drug cause analysis of AEs as well as the detection and interpretation of drug-related problems. Ongoing prospective evaluation of AEs and potential drug-related causes contributes to prevent patients from experiencing negative events. This process could be further accelerated by electronic assistance. Electronic documentation of AEs and computer-assisted signal detection of ADRs can support problem solving in a narrow timeframe since physicians and pharmacists are usually not permanently present in the LTC facilities [11, 30]. Database-supported comparison of the events with patients’ medication can assist pharmacists in a comprehensive medication review. Currently, such electronic solutions are rarely used in the LTC setting in Germany. They could also support future research by providing information on the additive effects of various combined drugs and underlying diseases. Thirdly, our data suggest the need for improvement in interdisciplinary communication in LTC facilities. In interprofessional teams with nurses, pharmacists and physicians, systematic information about AEs, medication reviews and actual health conditions could be transmitted more effectively in patient-orientated practice. Conclusion Nearly every long-term care resident suffered from adverse events (AEs), with half of them at least possibly caused by drugs. In four fifths of these AEs, several concomitantly given drugs were equally associated causes. Therefore, potential additive effects need to be considered independently from single causality and should be more focused in further research. A routinely implemented structured search for AEs and additive effects of polypharmacy contributes to medication reviews and interdisciplinary collaboration and will help to meet the needs of this complex patient collective and to protect them from negative consequences. Acknowledgements We thank all participating co-workers of the LTC facilities and the attending physicians for their support. Furthermore, we thank PhD Johanna Freyer for her assistance with the ethics approval and Katharine Worthington for language editing. Funding A co-worker of the study (Monika Lexow) was financially supported in part by the Lesmueller Foundation, Munich, Germany, the German Pharmacist Foundation, Berlin, Germany and the Pharmacist Foundation Westfalen-Lippe, Münster, Germany. Author Contribution Conceptualization: Monika Lexow, Kathrin Wernecke, Ralf Sulzer, Thilo Bertsche, Susanne Schiek; methodology: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; formal analysis and investigation: Kathrin Wernecke, Susanne Schiek; investigation: Monika Lexow, Kathrin Wernecke; writing, original draft preparation: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; writing, review and editing: Monika Lexow, Kathrin Wernecke, Ralf Sultzer, Gordian L. Schmid, Thilo Bertsche, Susanne Schiek; visualization: Monika Lexow, Kathrin Wernecke, Susanne Schiek; supervision: Thilo Bertsche, Susanne Schiek; project administration: Monika Lexow; funding acquisition: Thilo Bertsche, Susanne Schiek. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with ethical guidelines Conflict of interest M. Lexow, K. Wernecke, G.L. Schmid, R. Sultzer, T. Bertsche, and S. Schiek declare that they have no competing interests. Ethical standards All procedures performed in this study involving human participants were approved both by the ethics committee of the Faculty of Medicine of Leipzig University as well as the ethics committee of the State Chamber of Physicians of Saxony (reference: 231/13-ff and EK-allg-26/14-1) and have been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All participants gave their informed consent prior to inclusion in the study. The authors M. Lexow and K. Wernecke contributed equally to the manuscript. The authors T. Bertsche and S. Schiek contributed equally to the manuscript. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Oral
DrugAdministrationRoute
CC BY
33090261
20,066,723
2021-08
What was the dosage of drug 'DULOXETINE'?
Considering additive effects of polypharmacy : Analysis of adverse events in geriatric patients in long-term care facilities. BACKGROUND Potential additive effects of polypharmacy are rarely considered in adverse events of geriatric patients living in long-term care facilities. Our aim, therefore, was to identify adverse events in this setting and to assess plausible concomitant drug causes. METHODS A cross-sectional observational study was performed in three facilities as follows: (i) adverse event identification: we structurally identified adverse events using nurses' interviews and chart review. (ii) Analysis of the concomitantly administered drugs per patient was performed in two ways: (ii.a) a review of summary of product characteristics for listed adverse drug reactions to identify possible causing drugs and (ii.b) a causality assessment according to Naranjo algorithm. RESULTS (i) We found 424 adverse events with a median of 4 per patient (range 1-14) in 103 of the 104 enrolled patients (99%). (ii.a) We identified a median of 3 drugs (range 0-11) with actually occurring adverse events listed as an adverse drug reaction in the summary of product characteristics. (ii.b) Causality was classified in 198 (46.9%) of adverse events as "doubtful," in 218 (51.2%) as "possible," in 7 (1.7%) as "probable," and in 1 (0.2%) adverse event as a "definitive" cause of the administered drugs. In 340 (80.2%) of all identified adverse events several drugs simultaneously reached the highest respective Naranjo score. CONCLUSIONS Patients in long-term facilities frequently suffer from many adverse events. Concomitantly administered drugs have to be frequently considered as plausible causes for adverse events. These additive effects of drugs should be more focused in patient care and research. Introduction Geriatric patients in long-term care (LTC) facilities are multimorbid and, therefore, suffer from many (non)specific symptoms and geriatric syndromes [1]. Disease-related symptoms should be distinguished from adverse drug reactions (ADR) that result from drug therapy [2]. The latter can lead to hospital admissions and have a considerable impact on morbidity and mortality with high costs in the health care system [3–5]. Polypharmacy makes a significant contribution to the clinical consequences deriving from ADRs in geriatric patients [6]. For this reason, a structured analysis of adverse events (AE) and drug-related causes in these patients is of high interest for routine care. Distinguishing whether an observed AE was caused by a disease (i.e. symptom) or by a drug (ADR) poses a challenge for healthcare professionals [7, 8]. The correct attribution is required for appropriate treatment strategies but can result only from structured detection, analysis and classification. Geriatric patients are frequently cognitively impaired or suffer from speech or hearing disorders. Hence, information provided by the patients is often insufficient. In LTC facilities, therefore, chart documentation and nurses’ interviews are the most valuable sources for AE detection [9, 10]. So far, no specific method exists to analyze and classify AE in LTC facility residents with polypharmacy. The Naranjo algorithm has previously been used for causality assessment in this collective [11, 12]. It allows a detailed assessment of every detected AE and every single administered drug. This algorithm provides further information on drug-related causes when combined with established methods for patient safety, such as drug-drug interactions and potentially inappropriate medications [13]. Causality scores like the Naranjo score, however, do not consider simultaneously contributing drugs. For some ADRs, it has been shown that the number of specific drugs causes their clinical manifestations. For example, patients are exposed to an increased risk of falling when they take two or more drugs which increase the risk of falling [14]. Concerning anticholinergic ADRs, it is common to calculate an anticholinergic burden to quantify the risk for an adverse outcome [15]. Little is known, however, about additive drug effects in other events. Therefore, data about potential additive effects in this vulnerable patient collective are of great interest for routine care. The aim of this study was to identify AEs occurring in LTC facility patients and to assess plausible concomitant drug causes. Patients, material and methods Definitions We defined an AE as an outcome that occurs while a patient is taking a drug, but is not or not necessarily attributable to it and an ADR as an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product [16]. We used the term drug not only for the effective substance but for the whole product prescribed in the medication chart of the patient. A drug therefore could contain more than one active substance. We considered all drugs administered to the patient during the acquisition period. Continuous and on-demand medications were assessed separately because the temporal relationship between AE and administration of the drug could be different in that case. Participants and setting We conducted a cross-sectional observation study in three LTC facilities in Germany. After written informed consent of the residents or their legal representative and the responsible general practitioner, residents in the participating LTC facilities were enrolled in the study. We included residents of facilities with different ownerships (welfare, municipal or private associations) to approach a representative sample of 100 residents. Inclusion criteria were: informed consent, age ≥65 years, long-term/chronic medicines ≥3 and multimorbidity with ≥3 comorbidities at the time of recruitment, more than 8 weeks stay in the LTC facility, and a life expectancy of more than 6 months according to nurses’ present information. The study was conducted over a time period of 10 months. Study design and data collection We conducted a structured analysis of AEs. (i) AE identification We used two complement sources of information for our structured data collection: Firstly, an interview about individual AEs with nurses involved in daily care and, secondly, a review of residents’ records (electronic and chart documentation, laboratory values) for documented events and their temporal occurrence. To ensure standardized identification of AEs, a checklist of events was applied to both methods. The listed events comprised the most relevant AEs or ADRs for geriatric patients and LTC residents based on the literature [17–19]: blackened stool, bleeding/hematoma, confusion/disorientation, constipation, depression/anxiety, diarrhea, dizziness/vertigo, dry mouth, ear disorders, eye disorders, falls, hallucination, hyperglycemia/hypoglycemia, hyperhidrosis, hyperkalemia/hypokalemia, hypernatremia/hyponatremia, nausea, pain, restlessness, skin disorders/pruritus, insomnia, urinary incontinence, vomiting (in alphabetical order). Additional relevant reported or documented events were collected as well. We considered reported and documented symptoms during a time period of the prior 30 days (considered as 1 resident month) for new and continuous symptoms. Data collection was performed at two measurement points per patient at intervals of 6–8 weeks by two clinical pharmacists. All detected AEs and the corresponding system organ class were classified based on the common terminology criteria for adverse events (CTCAE) [20]. (iia) Review of summary of product characteristics We systematically collected data from the medical charts (continuous drugs, on demand drugs and their frequency of use, date of first administration). We checked all summaries of product characteristics (SmPCs) of the actually administered drugs for listed ADRs. A drug would be considered as “potentially causing” if the listed ADR in the SmPC represented a synonym for the detected AE or possibly caused it (e.g. dizziness in cases of falls). For the analysis of additive effects, we counted the number of potentially causing drugs. Prescribed drugs were characterized by their code in the anatomical therapeutic chemical classification system (ATC code). (iib) Causality assessment according to the Naranjo algorithm We used the Naranjo algorithm for causality assessment. All further relevant information, such as the duration of the AE, underlying diseases, clinical consequences (e.g. from hospital report), laboratory values, and patient-specific conditions were collected and used to determine the Naranjo score. The most likely associated drugs were the ones that reached the highest Naranjo score concerning the single analyzed AE. Naranjo distinguishes between definitive with a total score ≥9, probable with 5 < total score < 8, possible with 1 < total score < 4 and doubtful with a total score ≤ 0 [21, 22]. Inconclusive evaluations in all steps (i, ii.a, and ii.b) were discussed and finalized by mutual agreement in an expert panel. This panel consisted of four experienced clinical pharmacists. Statistical analysis To ensure comparable patient parameters between the three LTC facilities independent of the allocation to a single facility, main patient parameters were statistically analyzed. For this purpose, a Kruskal Wallis test with pairwise comparison was performed. Analyzed parameters were age, gender, number of diagnoses and number of continuous and on demand drugs, as well as the number of AEs in the patients and the maximum Naranjo score per patient. The data analysis was performed using IBM SPSS Statistics Version 25.0 (IBM Corporation, Armonk, NY, USA) and Microsoft Office Excel 2013 (Microsoft Corporation, Redmond, WA, USA). P-values ≤ 0.05 were considered as statistically significant. Results Patient characteristics In the participating parts of the LTC facilities, 182 patients were potentially available for the study and 154 met the inclusion criteria. From these, 104 patients or their legal guardian gave their informed consent as well as their responsible physician and were enrolled in the study. Patients were mostly female (72.1%) and in median 86 (range: 66–101) years old (Table 1). Patients did not differ between the three LTC facilities according to the following parameters: age (p = 0.311), gender (p = 0.684), number of diagnoses (p = 0.070) and number of continuous (p = 0.629) and on demand drugs (p = 0.911).Table 1 Characteristics of patients included in the study with frequency of documented diagnoses, main ATC classes and main active substances Characteristics Value Patients, total, n 104 Patients in facility of welfare ownership, n (%) 34 (32.7%) Patients in facility of municipal ownership, n (%) 30 (28.8%) Patients in facility of ownership by private association, n (%) 40 (38.5%) Female, n (%) 75 (72.1%) Length of residence (months), median (Q25/Q75; min–max) 31 (12/63; 1–414) Age (years), median (Q25/Q75; min–max) 86 (78/90; 66–101) Documented diagnoses, median (Q25/Q75; min–max) 15 (10/21; 3–35) No. of continuous drugs, median (Q25/Q75; min–max) 8 (6/10; 2–18) No. of on demand medication, median (Q25/Q75; min–max) 2 (1/3; 1–6) Documented diagnosisa Hypertension, n (%) 82 (78.8%) Dementia, n (%) 69 (66.3%) Diabetes, n (%) 41 (39.4%) Heart failure, n (%) 32 (30.8%) Atrial fibrillation, n (%) 32 (30.8%) Renal failure, n (%) 24 (23.1%) Osteoporosis, n (%) 19 (18.3%) Stroke, n (%) 17 (16.3%) Main ATC classesb C (cardiovascular system), n (%) 236 (28.7%) N (nervous system), n (%) 216 (26.3%) A (alimentary tract and metabolism), n (%) 164 (20.0%) B (blood and blood-forming organs), n (%) 68 (8.3%) H (systemic hormonal preparations, excluding sex hormones and insulins), n (%) 27 (3.3%) Main active substancesb Torasemide, n (%) 47 (5.7%) Pantoprazole, n (%) 40 (4.9%) Ramipril, n (%) 35 (4.3%) Acetylsalicylic acid, n (%) 33 (4.0%) Metoprolol, n (%) 23 (2.8%) ATC anatomical therapeutic chemical/defined daily dose classification, Q25/Q75 first and third quartile aOrder is based on the most relevant diagnoses found in literature data to geriatric patients bAccording to the documented continuous drugs (i) AE identification From a total of 104 patients, at least 1 AE was identified in 103 (99.0%). We identified 424 AEs, with a detected median of 4 (Q25/Q75: 2/5, range 1–14) AEs per patient, which equals 2.05 AEs per resident month. The identified AEs and the number of affected patients are shown in Table 2. The system organ classes renal and urinary disorder (87 patients), gastrointestinal disorder (43 patients), skin and subcutaneous tissue disorders (37 patients) were most common in our patient collective. Altogether, 72 different AE categories were detected, 185 AEs were identified in the patient records and 195 AEs by the nurses’ interviews, with 44 AEs in concordance of both methods. We found a significant difference in the detected number of AEs between the observed LTC facilities (p = 0.020). Following the pairwise comparison, we only found differences between the municipal LTC facility with 3 (Q25/Q75: 2/4) AEs and the private LTC facility with a median of 4 (Q25/Q75: 3/6.25) AEs (p = 0.022).Table 2 Identified adverse evnts (n = 424) according to CTCAE and affected patients (n = 104) System organ class Number of identified AEs, n (%) Affected patients, n (%) AE with number of affected patientsa (n) Renal and urinary disorders 88 (20.8) 87 (83.7) Urinary incontinence (87), urinary tract pain (1) Gastrointestinal disorders 55 (13.0) 43 (41.3) Constipation (22), vomiting (16), diarrhea (10), blackened stools (3), nausea (2), lower gastrointestinal bleeding (1), periodontal disease (1) Psychiatric disorders 55 (13.0) 35 (33.7) Confusion (21), restlessness (12), defensive behavior (8), insomnia (5), depression (4), anxiety (2), hallucinations (1), personality change (1), psychiatric disorders—other specify (1) Skin and subcutaneous tissue disorders 50 (11.8) 37 (35.6) Intertrigo (9), dry skin (7), hyperhidrosis (7), skin ulceration (6), local redness (5), pruritus (4), purpura (4), skin and subcutaneous tissue disorders—other specify (3), skin induration (1), urticaria (2), alopecia (1), angioedema (1) Metabolism and nutritional disorders 41 (9.7) 27 (26.0) Hyperglycemia (26), hypoglycemia (15) Musculoskeletal and connective tissue disorders 40 (9.4) 33 (31.7) Arthralgia (14), pain in extremity (12), back pain (6), arthritis (4), musculoskeletal and connective tissue disorders—other specify (3), general muscle weakness (1) Nervous system disorders 39 (9.2) 31 (29.8) Dizziness (11), somnolence (10), headache (4), syncope (3), ataxia (2), cognitive disturbance (2), paresthesia (2), depressed level of consciousness (1), lethargy (1), neuralgia (1), seizure (1), spasticity (1) Injury, poisoning and procedural complications 14 (3.3) 14 (13.5) Fall (14) Vascular disorders 11 (2.6) 11 (10.6) Hematoma (10), flushing (1) Infections and infestations 8 (1.9) 7 (6.7) Skin infection (4), vulval infection (2), conjunctivitis infective (1), stoma site infection (1) General disorders and administration site conditions 7 (1.7) 7 (6.7) Edema limbs (3), pain (3), fatigue (1) Respiratory, thoracic and mediastinal disorders 7 (1.7) 6 (5.8) Dyspnea (4), cough (1), epistaxis (1), respiratory, thoracic and mediastinal disorders—other specify (1) Ear and labyrinth disorders 3 (0.7) 3 (2.9) Hearing impaired (2), tinnitus (1) Cardiac disorders 2 (0.5) 2 (1.9) Chest pain—cardiac (1), palpitations (1) Eye disorders 2 (0.5) 1 (1.0) Blurred vision (1), glaucoma (1) Investigations 2 (0.5) 2 (1.9) Weight gain (1), weight loss (1) AE(s) adverse event(s), CTCAE common terminology criteria for adverse events aMultiple categories per patient possible (ii.a) Review of summary of product characteristics To analyze the concomitantly administered drugs, we assessed 3725 combinations of AEs and corresponding drugs. For this analysis five drug/AE pairs had to be excluded because no information from the SmPC was available (moisturizing eye drops, medical device). Considering every identified AE, patients had a median of 3 potentially causing drugs according to the SmPC, with a range from 0 to 11 drugs (Q25/Q75: 2/4; details in Fig. 1). The most frequently (n ≥ 10) detected AEs and the affected system organ classes are shown in Table 3. The ATC classes prescribed most often (C, N, A, B, H) were frequently among the potentially causing drugs for the most common system organ classes (Fig. 2).Fig. 1 Number of detected adverse events versus number of potentially causing drugs according Summary of Product Charactetistics. AE(s) adverse event(s), SmPC summary of products characteristics Table 3 Median number of potentially causing drugs according Summary of Product Charactersitics and corresponding Naranjo Score per patient (n = 104) for the most frequently detected (≥10) adverse events (AEs) and for their corresponding System organ classes (all 424 detected AE included) System organ class and ... most frequent AE Number (n) Median number of potentially causing drugs per patient [range] Median Naranjo score [range] Renal and urinary disorders 88 2 [0–5] 0 [−1–2] ... Urinary incontinence 87 2 [0–5] 0 [−1–2] Gastrointestinal disorders 55 5 [0–11] 2 [0–4] ... Constipation 22 5 [0–10] 0 [0–3] ... Vomiting 16 7.5 [2–10] 2 [0–4] ... Diarrhea 10 4.5 [2–11] 3 [0–3] Psychiatric disorders 55 3 [0–7] 0 [−1–9] ... Confusion 21 3 [1–7] 1 [0–8] ... Restlessness 12 3 [0–4] 0 [−1–3] Metabolism and nutrition disorders 41 3 [0–5] 1 [0–5] ... Hyperglycemia 26 3 [0–4] 1 [0–1] ... Hypoglycemia 15 3 [1–5] 4 [2–5] Musculoskeletal and connective tissue disorders 40 3 [0–8] 2 [−1–3] ... Arthralgia 14 3 [0–6] 1 [−1–3] ... Pain in extremity 12 2 [1–8] 2 [0–3] Nervous system disorders 39 4 [0–10] 1 [−1–7] ... Dizziness 11 5 [1–10] 2 [0–7] ... Somnolence 10 4 [3–7] 3 [0–5] Injury, poisoning and procedural complications 14 6 [3–11] 2 [0–3] ... Fall 14 6 [3–11] 2 [0–3] Vascular disorders 11 1 [0–3] 1 [0–3] ... Hematoma 10 1 [0–3] 0.5 [0–2] AE(s) Adverse Event(s), SmPC Summary of product characteristics Fig. 2 Potentially causing drugs according Summary of Product Characteristics differentiated into the ATC classes for the most frequently detected system organ classes. AE(s) adverse event(s), ATC anatomical therapeutic chemical/defined daily dose classification, CTCAE common criteria for the terminology of adverse events, SmPC summary of products characteristics. ATC classes: A: alimentary tract and metabolism, B: blood and blood forming organs, C: cardiovascular system, H: systemic hormonal preparations, excluding sex hormones and insulins, N: nervous system (ii.b) Causality assessment according to the Naranjo algorithm All 3730 drug/AE pairs were included in the causality assessment. From the 424 identified AEs, 198 (46.9%) were classified as ADR with “doubtful”, 218 (51.2%) “possible”, 7 (1.7%) “probable”, and 1 (0.2%) “definitive” cause (Table 4). We found no significant differences in the maximum Naranjo scores per patient between the three LTC facilities (p = 0.964). On the basis of 424 detected AEs, only 1 drug in 84 AEs (19.8%) and several drugs in 340 AEs (80.2%) reached the highest score (Table 4). According to Naranjo these need to be considered as the most likely causing drug(s).Table 4 Results of the adverse event drug causality assessment according to the Naranjo algorithm Naranjo Score per AE Identified AE [n] Number of affected patientsa, [n] Classification Identified AE per class, n (%) Number of AE with one/several highest Naranjo drug(s) [n] −1 22 17 Doubtful 199 (46.9) 38/161 0 177 92 1 68 48 Possible 217 (51.2) 38/179 2 90 51 3 41 30 4 18 18 5 3 3 Probable 7 (1.7) 7/0 6 2 2 7 1 1 8 1 1 9 1 1 Definitive 1 (0.2) 1/0 AE(s) adverse event(s) aSeveral AEs per patient possible The probable and definitive ADRs were as follows: angioedema (severity grade according to CTCAE 4) induced by enalapril, urticaria (grade 2) induced by amoxicillin and clavulanic acid, hypoglycemia (grade 1) induced by insulin glargine, paresthesia (grade 2) induced by tapentadol and a complex case of occurring hallucinations (grade 3) in combination with confusion (grade 3), dizziness (grade 3) and somnolence (grade 3, in total 4 detected AEs) which were attributable to digitoxin (highest Naranjo score). In this case, the patient also received high doses of oxycodone and duloxetine. It can be seen as a mixed intoxication based on the hospital report. In the algorithm, digitoxin reached a one-point higher score than oxycodone/duloxetine because measurement of the increased blood level was available only for digitoxin. All of the detected AEs and ADRs were managed adequately by the nurses, for example, by informing a physician or arranging a hospital admission for the affected patient. Thus, no further action was required due to this study. Discussion In our study we addressed AEs in geriatric patients living in LTC facilities. We assessed which type of AEs occurred and also investigated potential additive effects of polypharmacy. With nearly all (99%) patients affected by AEs, we demonstrated the relevance of this topic. We found the identified AEs potentially caused by up to 11 different administered drugs. The Naranjo algorithm showed at least possible drug causes in half of these AEs. Thereby, multiple drugs were equally likely involved 80% of the time. Our results point out that AEs should be systematically recorded in routine practice in LTC facilities. In order to prevent ADRs, additive effects need to be considered in any strategies developed. Prevalence of AE and ADR in LTC residents More than half of our identified AEs could be associated with drug use. Our rate of probable and definitive ADRs was similar to other studies in the LTC setting (0.04 vs. up to 0.10 ADRs per observed resident month), although studies should be compared with caution [9, 23]. Nevertheless, the causality assessment leaves us with a high number of possible ADRs. Especially for AEs which were ongoing for a longer period, causality assessment was challenging in the routine setting. Information to evaluate the exact temporal connection between AE and drug use was frequently missing and therefore could have led to lower Naranjo scores. To resolve this problem, a regular and structured routine assessment of AEs and potentially causing drugs might increase the chance to identify ADRs and protect patients from the consequences. Our overall rate of identified AEs was higher than results seen in other studies (2.05 AEs vs 0.03–0.12 per observed resident month) [9, 23]. This indicates that we identified a noticeable amount of the general symptom burden of LTC residents that results from underlying diseases or age-related changes. This is consistent with the fact that incontinence, pain, sleep disorders and psychopathological symptoms are widely found in LTC residents [24]. Therefore, a regular routine AE assessment can support ADR detection as well as structured symptom evaluation. Additive effects of polypharmacy The suspected AE was listed as an ADR in the respective SmPC in a median of 3 and up to 11 administered drugs per patient. In 80% of all identified AEs, various drugs reached the highest Naranjo score simultaneously. This means that they were equally likely to cause the AE. This coincidence can increase the chance of AE occurrence independently from single causality scores. This result also raises the question whether ADRs resulting from additive effects have been underestimated. In cases with “probable” or “definitive” ADRs (Naranjo ≥5), we found results from only one drug with the highest Naranjo score; however, in four of these AEs, the drug with the highest Naranjo (digitoxin) was only part of a mixed intoxication with duloxetine and oxycodone based on the hospital report for the affected patient. In this case, the sole consideration of the causality assessment could mask an additive effect of at least 3 concomitantly given drugs. This shows that additive effects need to be considered in every detected AE independently from the single causality. Besides ADRs from well-known drug classes (e.g. vascular ADRs from drugs affecting blood and blood-forming organs), in a substantial amount of AEs, we found involvement of varying ATC-classes that are less familiar (e.g. nervous system ADRs in drugs affecting the cardiovascular system). This underlines the complexity of geriatric patient treatment and the need for interdisciplinary medication reviews that include an assessment of drug-related problems, such as drug-drug interactions, potentially inappropriate medication, as well as ADRs [25, 26]. In routine care, however, potential additive effects are often not taken into account. In particular, new and unclear symptoms could be misinterpreted as new diseases and sometimes even lead to prescribing cascades [27, 28]. Implications for practice Firstly, our study shows the need for a good data base and a regular routine assessment of AEs that occur in LTC facilities. We found a very low concordance rate of only 10% between AEs detected in nurses’ interviews and those mentioned in the patient record analysis. This demonstrates the potential of information loss in LTC facilities due to heterogeneous and incomplete AE documentation [29]. It also indicates the potential of recall bias in the nurses. The identification of every occurring AE allows a better assessment of simultaneously occurring events. We found in our study, for example, a combination of vomiting and diarrhea that indicated an infection rather than an ADR. Furthermore, the information on patients’ current symptoms contributes to appropriate proposals for medication changes in cases of identified ADRs. Secondly, our results support the development of strategies with improved consideration of the additive effects of polypharmacy. Combining an AE assessment with structured medication reviews improves the drug cause analysis of AEs as well as the detection and interpretation of drug-related problems. Ongoing prospective evaluation of AEs and potential drug-related causes contributes to prevent patients from experiencing negative events. This process could be further accelerated by electronic assistance. Electronic documentation of AEs and computer-assisted signal detection of ADRs can support problem solving in a narrow timeframe since physicians and pharmacists are usually not permanently present in the LTC facilities [11, 30]. Database-supported comparison of the events with patients’ medication can assist pharmacists in a comprehensive medication review. Currently, such electronic solutions are rarely used in the LTC setting in Germany. They could also support future research by providing information on the additive effects of various combined drugs and underlying diseases. Thirdly, our data suggest the need for improvement in interdisciplinary communication in LTC facilities. In interprofessional teams with nurses, pharmacists and physicians, systematic information about AEs, medication reviews and actual health conditions could be transmitted more effectively in patient-orientated practice. Conclusion Nearly every long-term care resident suffered from adverse events (AEs), with half of them at least possibly caused by drugs. In four fifths of these AEs, several concomitantly given drugs were equally associated causes. Therefore, potential additive effects need to be considered independently from single causality and should be more focused in further research. A routinely implemented structured search for AEs and additive effects of polypharmacy contributes to medication reviews and interdisciplinary collaboration and will help to meet the needs of this complex patient collective and to protect them from negative consequences. Acknowledgements We thank all participating co-workers of the LTC facilities and the attending physicians for their support. Furthermore, we thank PhD Johanna Freyer for her assistance with the ethics approval and Katharine Worthington for language editing. Funding A co-worker of the study (Monika Lexow) was financially supported in part by the Lesmueller Foundation, Munich, Germany, the German Pharmacist Foundation, Berlin, Germany and the Pharmacist Foundation Westfalen-Lippe, Münster, Germany. Author Contribution Conceptualization: Monika Lexow, Kathrin Wernecke, Ralf Sulzer, Thilo Bertsche, Susanne Schiek; methodology: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; formal analysis and investigation: Kathrin Wernecke, Susanne Schiek; investigation: Monika Lexow, Kathrin Wernecke; writing, original draft preparation: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; writing, review and editing: Monika Lexow, Kathrin Wernecke, Ralf Sultzer, Gordian L. Schmid, Thilo Bertsche, Susanne Schiek; visualization: Monika Lexow, Kathrin Wernecke, Susanne Schiek; supervision: Thilo Bertsche, Susanne Schiek; project administration: Monika Lexow; funding acquisition: Thilo Bertsche, Susanne Schiek. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with ethical guidelines Conflict of interest M. Lexow, K. Wernecke, G.L. Schmid, R. Sultzer, T. Bertsche, and S. Schiek declare that they have no competing interests. Ethical standards All procedures performed in this study involving human participants were approved both by the ethics committee of the Faculty of Medicine of Leipzig University as well as the ethics committee of the State Chamber of Physicians of Saxony (reference: 231/13-ff and EK-allg-26/14-1) and have been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All participants gave their informed consent prior to inclusion in the study. The authors M. Lexow and K. Wernecke contributed equally to the manuscript. The authors T. Bertsche and S. Schiek contributed equally to the manuscript. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
UNK (HIGH DOSES OF DULOXETINE )
DrugDosageText
CC BY
33090261
20,066,723
2021-08
What was the dosage of drug 'OXYCODONE'?
Considering additive effects of polypharmacy : Analysis of adverse events in geriatric patients in long-term care facilities. BACKGROUND Potential additive effects of polypharmacy are rarely considered in adverse events of geriatric patients living in long-term care facilities. Our aim, therefore, was to identify adverse events in this setting and to assess plausible concomitant drug causes. METHODS A cross-sectional observational study was performed in three facilities as follows: (i) adverse event identification: we structurally identified adverse events using nurses' interviews and chart review. (ii) Analysis of the concomitantly administered drugs per patient was performed in two ways: (ii.a) a review of summary of product characteristics for listed adverse drug reactions to identify possible causing drugs and (ii.b) a causality assessment according to Naranjo algorithm. RESULTS (i) We found 424 adverse events with a median of 4 per patient (range 1-14) in 103 of the 104 enrolled patients (99%). (ii.a) We identified a median of 3 drugs (range 0-11) with actually occurring adverse events listed as an adverse drug reaction in the summary of product characteristics. (ii.b) Causality was classified in 198 (46.9%) of adverse events as "doubtful," in 218 (51.2%) as "possible," in 7 (1.7%) as "probable," and in 1 (0.2%) adverse event as a "definitive" cause of the administered drugs. In 340 (80.2%) of all identified adverse events several drugs simultaneously reached the highest respective Naranjo score. CONCLUSIONS Patients in long-term facilities frequently suffer from many adverse events. Concomitantly administered drugs have to be frequently considered as plausible causes for adverse events. These additive effects of drugs should be more focused in patient care and research. Introduction Geriatric patients in long-term care (LTC) facilities are multimorbid and, therefore, suffer from many (non)specific symptoms and geriatric syndromes [1]. Disease-related symptoms should be distinguished from adverse drug reactions (ADR) that result from drug therapy [2]. The latter can lead to hospital admissions and have a considerable impact on morbidity and mortality with high costs in the health care system [3–5]. Polypharmacy makes a significant contribution to the clinical consequences deriving from ADRs in geriatric patients [6]. For this reason, a structured analysis of adverse events (AE) and drug-related causes in these patients is of high interest for routine care. Distinguishing whether an observed AE was caused by a disease (i.e. symptom) or by a drug (ADR) poses a challenge for healthcare professionals [7, 8]. The correct attribution is required for appropriate treatment strategies but can result only from structured detection, analysis and classification. Geriatric patients are frequently cognitively impaired or suffer from speech or hearing disorders. Hence, information provided by the patients is often insufficient. In LTC facilities, therefore, chart documentation and nurses’ interviews are the most valuable sources for AE detection [9, 10]. So far, no specific method exists to analyze and classify AE in LTC facility residents with polypharmacy. The Naranjo algorithm has previously been used for causality assessment in this collective [11, 12]. It allows a detailed assessment of every detected AE and every single administered drug. This algorithm provides further information on drug-related causes when combined with established methods for patient safety, such as drug-drug interactions and potentially inappropriate medications [13]. Causality scores like the Naranjo score, however, do not consider simultaneously contributing drugs. For some ADRs, it has been shown that the number of specific drugs causes their clinical manifestations. For example, patients are exposed to an increased risk of falling when they take two or more drugs which increase the risk of falling [14]. Concerning anticholinergic ADRs, it is common to calculate an anticholinergic burden to quantify the risk for an adverse outcome [15]. Little is known, however, about additive drug effects in other events. Therefore, data about potential additive effects in this vulnerable patient collective are of great interest for routine care. The aim of this study was to identify AEs occurring in LTC facility patients and to assess plausible concomitant drug causes. Patients, material and methods Definitions We defined an AE as an outcome that occurs while a patient is taking a drug, but is not or not necessarily attributable to it and an ADR as an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product [16]. We used the term drug not only for the effective substance but for the whole product prescribed in the medication chart of the patient. A drug therefore could contain more than one active substance. We considered all drugs administered to the patient during the acquisition period. Continuous and on-demand medications were assessed separately because the temporal relationship between AE and administration of the drug could be different in that case. Participants and setting We conducted a cross-sectional observation study in three LTC facilities in Germany. After written informed consent of the residents or their legal representative and the responsible general practitioner, residents in the participating LTC facilities were enrolled in the study. We included residents of facilities with different ownerships (welfare, municipal or private associations) to approach a representative sample of 100 residents. Inclusion criteria were: informed consent, age ≥65 years, long-term/chronic medicines ≥3 and multimorbidity with ≥3 comorbidities at the time of recruitment, more than 8 weeks stay in the LTC facility, and a life expectancy of more than 6 months according to nurses’ present information. The study was conducted over a time period of 10 months. Study design and data collection We conducted a structured analysis of AEs. (i) AE identification We used two complement sources of information for our structured data collection: Firstly, an interview about individual AEs with nurses involved in daily care and, secondly, a review of residents’ records (electronic and chart documentation, laboratory values) for documented events and their temporal occurrence. To ensure standardized identification of AEs, a checklist of events was applied to both methods. The listed events comprised the most relevant AEs or ADRs for geriatric patients and LTC residents based on the literature [17–19]: blackened stool, bleeding/hematoma, confusion/disorientation, constipation, depression/anxiety, diarrhea, dizziness/vertigo, dry mouth, ear disorders, eye disorders, falls, hallucination, hyperglycemia/hypoglycemia, hyperhidrosis, hyperkalemia/hypokalemia, hypernatremia/hyponatremia, nausea, pain, restlessness, skin disorders/pruritus, insomnia, urinary incontinence, vomiting (in alphabetical order). Additional relevant reported or documented events were collected as well. We considered reported and documented symptoms during a time period of the prior 30 days (considered as 1 resident month) for new and continuous symptoms. Data collection was performed at two measurement points per patient at intervals of 6–8 weeks by two clinical pharmacists. All detected AEs and the corresponding system organ class were classified based on the common terminology criteria for adverse events (CTCAE) [20]. (iia) Review of summary of product characteristics We systematically collected data from the medical charts (continuous drugs, on demand drugs and their frequency of use, date of first administration). We checked all summaries of product characteristics (SmPCs) of the actually administered drugs for listed ADRs. A drug would be considered as “potentially causing” if the listed ADR in the SmPC represented a synonym for the detected AE or possibly caused it (e.g. dizziness in cases of falls). For the analysis of additive effects, we counted the number of potentially causing drugs. Prescribed drugs were characterized by their code in the anatomical therapeutic chemical classification system (ATC code). (iib) Causality assessment according to the Naranjo algorithm We used the Naranjo algorithm for causality assessment. All further relevant information, such as the duration of the AE, underlying diseases, clinical consequences (e.g. from hospital report), laboratory values, and patient-specific conditions were collected and used to determine the Naranjo score. The most likely associated drugs were the ones that reached the highest Naranjo score concerning the single analyzed AE. Naranjo distinguishes between definitive with a total score ≥9, probable with 5 < total score < 8, possible with 1 < total score < 4 and doubtful with a total score ≤ 0 [21, 22]. Inconclusive evaluations in all steps (i, ii.a, and ii.b) were discussed and finalized by mutual agreement in an expert panel. This panel consisted of four experienced clinical pharmacists. Statistical analysis To ensure comparable patient parameters between the three LTC facilities independent of the allocation to a single facility, main patient parameters were statistically analyzed. For this purpose, a Kruskal Wallis test with pairwise comparison was performed. Analyzed parameters were age, gender, number of diagnoses and number of continuous and on demand drugs, as well as the number of AEs in the patients and the maximum Naranjo score per patient. The data analysis was performed using IBM SPSS Statistics Version 25.0 (IBM Corporation, Armonk, NY, USA) and Microsoft Office Excel 2013 (Microsoft Corporation, Redmond, WA, USA). P-values ≤ 0.05 were considered as statistically significant. Results Patient characteristics In the participating parts of the LTC facilities, 182 patients were potentially available for the study and 154 met the inclusion criteria. From these, 104 patients or their legal guardian gave their informed consent as well as their responsible physician and were enrolled in the study. Patients were mostly female (72.1%) and in median 86 (range: 66–101) years old (Table 1). Patients did not differ between the three LTC facilities according to the following parameters: age (p = 0.311), gender (p = 0.684), number of diagnoses (p = 0.070) and number of continuous (p = 0.629) and on demand drugs (p = 0.911).Table 1 Characteristics of patients included in the study with frequency of documented diagnoses, main ATC classes and main active substances Characteristics Value Patients, total, n 104 Patients in facility of welfare ownership, n (%) 34 (32.7%) Patients in facility of municipal ownership, n (%) 30 (28.8%) Patients in facility of ownership by private association, n (%) 40 (38.5%) Female, n (%) 75 (72.1%) Length of residence (months), median (Q25/Q75; min–max) 31 (12/63; 1–414) Age (years), median (Q25/Q75; min–max) 86 (78/90; 66–101) Documented diagnoses, median (Q25/Q75; min–max) 15 (10/21; 3–35) No. of continuous drugs, median (Q25/Q75; min–max) 8 (6/10; 2–18) No. of on demand medication, median (Q25/Q75; min–max) 2 (1/3; 1–6) Documented diagnosisa Hypertension, n (%) 82 (78.8%) Dementia, n (%) 69 (66.3%) Diabetes, n (%) 41 (39.4%) Heart failure, n (%) 32 (30.8%) Atrial fibrillation, n (%) 32 (30.8%) Renal failure, n (%) 24 (23.1%) Osteoporosis, n (%) 19 (18.3%) Stroke, n (%) 17 (16.3%) Main ATC classesb C (cardiovascular system), n (%) 236 (28.7%) N (nervous system), n (%) 216 (26.3%) A (alimentary tract and metabolism), n (%) 164 (20.0%) B (blood and blood-forming organs), n (%) 68 (8.3%) H (systemic hormonal preparations, excluding sex hormones and insulins), n (%) 27 (3.3%) Main active substancesb Torasemide, n (%) 47 (5.7%) Pantoprazole, n (%) 40 (4.9%) Ramipril, n (%) 35 (4.3%) Acetylsalicylic acid, n (%) 33 (4.0%) Metoprolol, n (%) 23 (2.8%) ATC anatomical therapeutic chemical/defined daily dose classification, Q25/Q75 first and third quartile aOrder is based on the most relevant diagnoses found in literature data to geriatric patients bAccording to the documented continuous drugs (i) AE identification From a total of 104 patients, at least 1 AE was identified in 103 (99.0%). We identified 424 AEs, with a detected median of 4 (Q25/Q75: 2/5, range 1–14) AEs per patient, which equals 2.05 AEs per resident month. The identified AEs and the number of affected patients are shown in Table 2. The system organ classes renal and urinary disorder (87 patients), gastrointestinal disorder (43 patients), skin and subcutaneous tissue disorders (37 patients) were most common in our patient collective. Altogether, 72 different AE categories were detected, 185 AEs were identified in the patient records and 195 AEs by the nurses’ interviews, with 44 AEs in concordance of both methods. We found a significant difference in the detected number of AEs between the observed LTC facilities (p = 0.020). Following the pairwise comparison, we only found differences between the municipal LTC facility with 3 (Q25/Q75: 2/4) AEs and the private LTC facility with a median of 4 (Q25/Q75: 3/6.25) AEs (p = 0.022).Table 2 Identified adverse evnts (n = 424) according to CTCAE and affected patients (n = 104) System organ class Number of identified AEs, n (%) Affected patients, n (%) AE with number of affected patientsa (n) Renal and urinary disorders 88 (20.8) 87 (83.7) Urinary incontinence (87), urinary tract pain (1) Gastrointestinal disorders 55 (13.0) 43 (41.3) Constipation (22), vomiting (16), diarrhea (10), blackened stools (3), nausea (2), lower gastrointestinal bleeding (1), periodontal disease (1) Psychiatric disorders 55 (13.0) 35 (33.7) Confusion (21), restlessness (12), defensive behavior (8), insomnia (5), depression (4), anxiety (2), hallucinations (1), personality change (1), psychiatric disorders—other specify (1) Skin and subcutaneous tissue disorders 50 (11.8) 37 (35.6) Intertrigo (9), dry skin (7), hyperhidrosis (7), skin ulceration (6), local redness (5), pruritus (4), purpura (4), skin and subcutaneous tissue disorders—other specify (3), skin induration (1), urticaria (2), alopecia (1), angioedema (1) Metabolism and nutritional disorders 41 (9.7) 27 (26.0) Hyperglycemia (26), hypoglycemia (15) Musculoskeletal and connective tissue disorders 40 (9.4) 33 (31.7) Arthralgia (14), pain in extremity (12), back pain (6), arthritis (4), musculoskeletal and connective tissue disorders—other specify (3), general muscle weakness (1) Nervous system disorders 39 (9.2) 31 (29.8) Dizziness (11), somnolence (10), headache (4), syncope (3), ataxia (2), cognitive disturbance (2), paresthesia (2), depressed level of consciousness (1), lethargy (1), neuralgia (1), seizure (1), spasticity (1) Injury, poisoning and procedural complications 14 (3.3) 14 (13.5) Fall (14) Vascular disorders 11 (2.6) 11 (10.6) Hematoma (10), flushing (1) Infections and infestations 8 (1.9) 7 (6.7) Skin infection (4), vulval infection (2), conjunctivitis infective (1), stoma site infection (1) General disorders and administration site conditions 7 (1.7) 7 (6.7) Edema limbs (3), pain (3), fatigue (1) Respiratory, thoracic and mediastinal disorders 7 (1.7) 6 (5.8) Dyspnea (4), cough (1), epistaxis (1), respiratory, thoracic and mediastinal disorders—other specify (1) Ear and labyrinth disorders 3 (0.7) 3 (2.9) Hearing impaired (2), tinnitus (1) Cardiac disorders 2 (0.5) 2 (1.9) Chest pain—cardiac (1), palpitations (1) Eye disorders 2 (0.5) 1 (1.0) Blurred vision (1), glaucoma (1) Investigations 2 (0.5) 2 (1.9) Weight gain (1), weight loss (1) AE(s) adverse event(s), CTCAE common terminology criteria for adverse events aMultiple categories per patient possible (ii.a) Review of summary of product characteristics To analyze the concomitantly administered drugs, we assessed 3725 combinations of AEs and corresponding drugs. For this analysis five drug/AE pairs had to be excluded because no information from the SmPC was available (moisturizing eye drops, medical device). Considering every identified AE, patients had a median of 3 potentially causing drugs according to the SmPC, with a range from 0 to 11 drugs (Q25/Q75: 2/4; details in Fig. 1). The most frequently (n ≥ 10) detected AEs and the affected system organ classes are shown in Table 3. The ATC classes prescribed most often (C, N, A, B, H) were frequently among the potentially causing drugs for the most common system organ classes (Fig. 2).Fig. 1 Number of detected adverse events versus number of potentially causing drugs according Summary of Product Charactetistics. AE(s) adverse event(s), SmPC summary of products characteristics Table 3 Median number of potentially causing drugs according Summary of Product Charactersitics and corresponding Naranjo Score per patient (n = 104) for the most frequently detected (≥10) adverse events (AEs) and for their corresponding System organ classes (all 424 detected AE included) System organ class and ... most frequent AE Number (n) Median number of potentially causing drugs per patient [range] Median Naranjo score [range] Renal and urinary disorders 88 2 [0–5] 0 [−1–2] ... Urinary incontinence 87 2 [0–5] 0 [−1–2] Gastrointestinal disorders 55 5 [0–11] 2 [0–4] ... Constipation 22 5 [0–10] 0 [0–3] ... Vomiting 16 7.5 [2–10] 2 [0–4] ... Diarrhea 10 4.5 [2–11] 3 [0–3] Psychiatric disorders 55 3 [0–7] 0 [−1–9] ... Confusion 21 3 [1–7] 1 [0–8] ... Restlessness 12 3 [0–4] 0 [−1–3] Metabolism and nutrition disorders 41 3 [0–5] 1 [0–5] ... Hyperglycemia 26 3 [0–4] 1 [0–1] ... Hypoglycemia 15 3 [1–5] 4 [2–5] Musculoskeletal and connective tissue disorders 40 3 [0–8] 2 [−1–3] ... Arthralgia 14 3 [0–6] 1 [−1–3] ... Pain in extremity 12 2 [1–8] 2 [0–3] Nervous system disorders 39 4 [0–10] 1 [−1–7] ... Dizziness 11 5 [1–10] 2 [0–7] ... Somnolence 10 4 [3–7] 3 [0–5] Injury, poisoning and procedural complications 14 6 [3–11] 2 [0–3] ... Fall 14 6 [3–11] 2 [0–3] Vascular disorders 11 1 [0–3] 1 [0–3] ... Hematoma 10 1 [0–3] 0.5 [0–2] AE(s) Adverse Event(s), SmPC Summary of product characteristics Fig. 2 Potentially causing drugs according Summary of Product Characteristics differentiated into the ATC classes for the most frequently detected system organ classes. AE(s) adverse event(s), ATC anatomical therapeutic chemical/defined daily dose classification, CTCAE common criteria for the terminology of adverse events, SmPC summary of products characteristics. ATC classes: A: alimentary tract and metabolism, B: blood and blood forming organs, C: cardiovascular system, H: systemic hormonal preparations, excluding sex hormones and insulins, N: nervous system (ii.b) Causality assessment according to the Naranjo algorithm All 3730 drug/AE pairs were included in the causality assessment. From the 424 identified AEs, 198 (46.9%) were classified as ADR with “doubtful”, 218 (51.2%) “possible”, 7 (1.7%) “probable”, and 1 (0.2%) “definitive” cause (Table 4). We found no significant differences in the maximum Naranjo scores per patient between the three LTC facilities (p = 0.964). On the basis of 424 detected AEs, only 1 drug in 84 AEs (19.8%) and several drugs in 340 AEs (80.2%) reached the highest score (Table 4). According to Naranjo these need to be considered as the most likely causing drug(s).Table 4 Results of the adverse event drug causality assessment according to the Naranjo algorithm Naranjo Score per AE Identified AE [n] Number of affected patientsa, [n] Classification Identified AE per class, n (%) Number of AE with one/several highest Naranjo drug(s) [n] −1 22 17 Doubtful 199 (46.9) 38/161 0 177 92 1 68 48 Possible 217 (51.2) 38/179 2 90 51 3 41 30 4 18 18 5 3 3 Probable 7 (1.7) 7/0 6 2 2 7 1 1 8 1 1 9 1 1 Definitive 1 (0.2) 1/0 AE(s) adverse event(s) aSeveral AEs per patient possible The probable and definitive ADRs were as follows: angioedema (severity grade according to CTCAE 4) induced by enalapril, urticaria (grade 2) induced by amoxicillin and clavulanic acid, hypoglycemia (grade 1) induced by insulin glargine, paresthesia (grade 2) induced by tapentadol and a complex case of occurring hallucinations (grade 3) in combination with confusion (grade 3), dizziness (grade 3) and somnolence (grade 3, in total 4 detected AEs) which were attributable to digitoxin (highest Naranjo score). In this case, the patient also received high doses of oxycodone and duloxetine. It can be seen as a mixed intoxication based on the hospital report. In the algorithm, digitoxin reached a one-point higher score than oxycodone/duloxetine because measurement of the increased blood level was available only for digitoxin. All of the detected AEs and ADRs were managed adequately by the nurses, for example, by informing a physician or arranging a hospital admission for the affected patient. Thus, no further action was required due to this study. Discussion In our study we addressed AEs in geriatric patients living in LTC facilities. We assessed which type of AEs occurred and also investigated potential additive effects of polypharmacy. With nearly all (99%) patients affected by AEs, we demonstrated the relevance of this topic. We found the identified AEs potentially caused by up to 11 different administered drugs. The Naranjo algorithm showed at least possible drug causes in half of these AEs. Thereby, multiple drugs were equally likely involved 80% of the time. Our results point out that AEs should be systematically recorded in routine practice in LTC facilities. In order to prevent ADRs, additive effects need to be considered in any strategies developed. Prevalence of AE and ADR in LTC residents More than half of our identified AEs could be associated with drug use. Our rate of probable and definitive ADRs was similar to other studies in the LTC setting (0.04 vs. up to 0.10 ADRs per observed resident month), although studies should be compared with caution [9, 23]. Nevertheless, the causality assessment leaves us with a high number of possible ADRs. Especially for AEs which were ongoing for a longer period, causality assessment was challenging in the routine setting. Information to evaluate the exact temporal connection between AE and drug use was frequently missing and therefore could have led to lower Naranjo scores. To resolve this problem, a regular and structured routine assessment of AEs and potentially causing drugs might increase the chance to identify ADRs and protect patients from the consequences. Our overall rate of identified AEs was higher than results seen in other studies (2.05 AEs vs 0.03–0.12 per observed resident month) [9, 23]. This indicates that we identified a noticeable amount of the general symptom burden of LTC residents that results from underlying diseases or age-related changes. This is consistent with the fact that incontinence, pain, sleep disorders and psychopathological symptoms are widely found in LTC residents [24]. Therefore, a regular routine AE assessment can support ADR detection as well as structured symptom evaluation. Additive effects of polypharmacy The suspected AE was listed as an ADR in the respective SmPC in a median of 3 and up to 11 administered drugs per patient. In 80% of all identified AEs, various drugs reached the highest Naranjo score simultaneously. This means that they were equally likely to cause the AE. This coincidence can increase the chance of AE occurrence independently from single causality scores. This result also raises the question whether ADRs resulting from additive effects have been underestimated. In cases with “probable” or “definitive” ADRs (Naranjo ≥5), we found results from only one drug with the highest Naranjo score; however, in four of these AEs, the drug with the highest Naranjo (digitoxin) was only part of a mixed intoxication with duloxetine and oxycodone based on the hospital report for the affected patient. In this case, the sole consideration of the causality assessment could mask an additive effect of at least 3 concomitantly given drugs. This shows that additive effects need to be considered in every detected AE independently from the single causality. Besides ADRs from well-known drug classes (e.g. vascular ADRs from drugs affecting blood and blood-forming organs), in a substantial amount of AEs, we found involvement of varying ATC-classes that are less familiar (e.g. nervous system ADRs in drugs affecting the cardiovascular system). This underlines the complexity of geriatric patient treatment and the need for interdisciplinary medication reviews that include an assessment of drug-related problems, such as drug-drug interactions, potentially inappropriate medication, as well as ADRs [25, 26]. In routine care, however, potential additive effects are often not taken into account. In particular, new and unclear symptoms could be misinterpreted as new diseases and sometimes even lead to prescribing cascades [27, 28]. Implications for practice Firstly, our study shows the need for a good data base and a regular routine assessment of AEs that occur in LTC facilities. We found a very low concordance rate of only 10% between AEs detected in nurses’ interviews and those mentioned in the patient record analysis. This demonstrates the potential of information loss in LTC facilities due to heterogeneous and incomplete AE documentation [29]. It also indicates the potential of recall bias in the nurses. The identification of every occurring AE allows a better assessment of simultaneously occurring events. We found in our study, for example, a combination of vomiting and diarrhea that indicated an infection rather than an ADR. Furthermore, the information on patients’ current symptoms contributes to appropriate proposals for medication changes in cases of identified ADRs. Secondly, our results support the development of strategies with improved consideration of the additive effects of polypharmacy. Combining an AE assessment with structured medication reviews improves the drug cause analysis of AEs as well as the detection and interpretation of drug-related problems. Ongoing prospective evaluation of AEs and potential drug-related causes contributes to prevent patients from experiencing negative events. This process could be further accelerated by electronic assistance. Electronic documentation of AEs and computer-assisted signal detection of ADRs can support problem solving in a narrow timeframe since physicians and pharmacists are usually not permanently present in the LTC facilities [11, 30]. Database-supported comparison of the events with patients’ medication can assist pharmacists in a comprehensive medication review. Currently, such electronic solutions are rarely used in the LTC setting in Germany. They could also support future research by providing information on the additive effects of various combined drugs and underlying diseases. Thirdly, our data suggest the need for improvement in interdisciplinary communication in LTC facilities. In interprofessional teams with nurses, pharmacists and physicians, systematic information about AEs, medication reviews and actual health conditions could be transmitted more effectively in patient-orientated practice. Conclusion Nearly every long-term care resident suffered from adverse events (AEs), with half of them at least possibly caused by drugs. In four fifths of these AEs, several concomitantly given drugs were equally associated causes. Therefore, potential additive effects need to be considered independently from single causality and should be more focused in further research. A routinely implemented structured search for AEs and additive effects of polypharmacy contributes to medication reviews and interdisciplinary collaboration and will help to meet the needs of this complex patient collective and to protect them from negative consequences. Acknowledgements We thank all participating co-workers of the LTC facilities and the attending physicians for their support. Furthermore, we thank PhD Johanna Freyer for her assistance with the ethics approval and Katharine Worthington for language editing. Funding A co-worker of the study (Monika Lexow) was financially supported in part by the Lesmueller Foundation, Munich, Germany, the German Pharmacist Foundation, Berlin, Germany and the Pharmacist Foundation Westfalen-Lippe, Münster, Germany. Author Contribution Conceptualization: Monika Lexow, Kathrin Wernecke, Ralf Sulzer, Thilo Bertsche, Susanne Schiek; methodology: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; formal analysis and investigation: Kathrin Wernecke, Susanne Schiek; investigation: Monika Lexow, Kathrin Wernecke; writing, original draft preparation: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; writing, review and editing: Monika Lexow, Kathrin Wernecke, Ralf Sultzer, Gordian L. Schmid, Thilo Bertsche, Susanne Schiek; visualization: Monika Lexow, Kathrin Wernecke, Susanne Schiek; supervision: Thilo Bertsche, Susanne Schiek; project administration: Monika Lexow; funding acquisition: Thilo Bertsche, Susanne Schiek. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with ethical guidelines Conflict of interest M. Lexow, K. Wernecke, G.L. Schmid, R. Sultzer, T. Bertsche, and S. Schiek declare that they have no competing interests. Ethical standards All procedures performed in this study involving human participants were approved both by the ethics committee of the Faculty of Medicine of Leipzig University as well as the ethics committee of the State Chamber of Physicians of Saxony (reference: 231/13-ff and EK-allg-26/14-1) and have been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All participants gave their informed consent prior to inclusion in the study. The authors M. Lexow and K. Wernecke contributed equally to the manuscript. The authors T. Bertsche and S. Schiek contributed equally to the manuscript. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
UNK (HIGH DOSES OF OXYCODONE)
DrugDosageText
CC BY
33090261
20,066,723
2021-08
What was the outcome of reaction 'Confusional state'?
Considering additive effects of polypharmacy : Analysis of adverse events in geriatric patients in long-term care facilities. BACKGROUND Potential additive effects of polypharmacy are rarely considered in adverse events of geriatric patients living in long-term care facilities. Our aim, therefore, was to identify adverse events in this setting and to assess plausible concomitant drug causes. METHODS A cross-sectional observational study was performed in three facilities as follows: (i) adverse event identification: we structurally identified adverse events using nurses' interviews and chart review. (ii) Analysis of the concomitantly administered drugs per patient was performed in two ways: (ii.a) a review of summary of product characteristics for listed adverse drug reactions to identify possible causing drugs and (ii.b) a causality assessment according to Naranjo algorithm. RESULTS (i) We found 424 adverse events with a median of 4 per patient (range 1-14) in 103 of the 104 enrolled patients (99%). (ii.a) We identified a median of 3 drugs (range 0-11) with actually occurring adverse events listed as an adverse drug reaction in the summary of product characteristics. (ii.b) Causality was classified in 198 (46.9%) of adverse events as "doubtful," in 218 (51.2%) as "possible," in 7 (1.7%) as "probable," and in 1 (0.2%) adverse event as a "definitive" cause of the administered drugs. In 340 (80.2%) of all identified adverse events several drugs simultaneously reached the highest respective Naranjo score. CONCLUSIONS Patients in long-term facilities frequently suffer from many adverse events. Concomitantly administered drugs have to be frequently considered as plausible causes for adverse events. These additive effects of drugs should be more focused in patient care and research. Introduction Geriatric patients in long-term care (LTC) facilities are multimorbid and, therefore, suffer from many (non)specific symptoms and geriatric syndromes [1]. Disease-related symptoms should be distinguished from adverse drug reactions (ADR) that result from drug therapy [2]. The latter can lead to hospital admissions and have a considerable impact on morbidity and mortality with high costs in the health care system [3–5]. Polypharmacy makes a significant contribution to the clinical consequences deriving from ADRs in geriatric patients [6]. For this reason, a structured analysis of adverse events (AE) and drug-related causes in these patients is of high interest for routine care. Distinguishing whether an observed AE was caused by a disease (i.e. symptom) or by a drug (ADR) poses a challenge for healthcare professionals [7, 8]. The correct attribution is required for appropriate treatment strategies but can result only from structured detection, analysis and classification. Geriatric patients are frequently cognitively impaired or suffer from speech or hearing disorders. Hence, information provided by the patients is often insufficient. In LTC facilities, therefore, chart documentation and nurses’ interviews are the most valuable sources for AE detection [9, 10]. So far, no specific method exists to analyze and classify AE in LTC facility residents with polypharmacy. The Naranjo algorithm has previously been used for causality assessment in this collective [11, 12]. It allows a detailed assessment of every detected AE and every single administered drug. This algorithm provides further information on drug-related causes when combined with established methods for patient safety, such as drug-drug interactions and potentially inappropriate medications [13]. Causality scores like the Naranjo score, however, do not consider simultaneously contributing drugs. For some ADRs, it has been shown that the number of specific drugs causes their clinical manifestations. For example, patients are exposed to an increased risk of falling when they take two or more drugs which increase the risk of falling [14]. Concerning anticholinergic ADRs, it is common to calculate an anticholinergic burden to quantify the risk for an adverse outcome [15]. Little is known, however, about additive drug effects in other events. Therefore, data about potential additive effects in this vulnerable patient collective are of great interest for routine care. The aim of this study was to identify AEs occurring in LTC facility patients and to assess plausible concomitant drug causes. Patients, material and methods Definitions We defined an AE as an outcome that occurs while a patient is taking a drug, but is not or not necessarily attributable to it and an ADR as an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product [16]. We used the term drug not only for the effective substance but for the whole product prescribed in the medication chart of the patient. A drug therefore could contain more than one active substance. We considered all drugs administered to the patient during the acquisition period. Continuous and on-demand medications were assessed separately because the temporal relationship between AE and administration of the drug could be different in that case. Participants and setting We conducted a cross-sectional observation study in three LTC facilities in Germany. After written informed consent of the residents or their legal representative and the responsible general practitioner, residents in the participating LTC facilities were enrolled in the study. We included residents of facilities with different ownerships (welfare, municipal or private associations) to approach a representative sample of 100 residents. Inclusion criteria were: informed consent, age ≥65 years, long-term/chronic medicines ≥3 and multimorbidity with ≥3 comorbidities at the time of recruitment, more than 8 weeks stay in the LTC facility, and a life expectancy of more than 6 months according to nurses’ present information. The study was conducted over a time period of 10 months. Study design and data collection We conducted a structured analysis of AEs. (i) AE identification We used two complement sources of information for our structured data collection: Firstly, an interview about individual AEs with nurses involved in daily care and, secondly, a review of residents’ records (electronic and chart documentation, laboratory values) for documented events and their temporal occurrence. To ensure standardized identification of AEs, a checklist of events was applied to both methods. The listed events comprised the most relevant AEs or ADRs for geriatric patients and LTC residents based on the literature [17–19]: blackened stool, bleeding/hematoma, confusion/disorientation, constipation, depression/anxiety, diarrhea, dizziness/vertigo, dry mouth, ear disorders, eye disorders, falls, hallucination, hyperglycemia/hypoglycemia, hyperhidrosis, hyperkalemia/hypokalemia, hypernatremia/hyponatremia, nausea, pain, restlessness, skin disorders/pruritus, insomnia, urinary incontinence, vomiting (in alphabetical order). Additional relevant reported or documented events were collected as well. We considered reported and documented symptoms during a time period of the prior 30 days (considered as 1 resident month) for new and continuous symptoms. Data collection was performed at two measurement points per patient at intervals of 6–8 weeks by two clinical pharmacists. All detected AEs and the corresponding system organ class were classified based on the common terminology criteria for adverse events (CTCAE) [20]. (iia) Review of summary of product characteristics We systematically collected data from the medical charts (continuous drugs, on demand drugs and their frequency of use, date of first administration). We checked all summaries of product characteristics (SmPCs) of the actually administered drugs for listed ADRs. A drug would be considered as “potentially causing” if the listed ADR in the SmPC represented a synonym for the detected AE or possibly caused it (e.g. dizziness in cases of falls). For the analysis of additive effects, we counted the number of potentially causing drugs. Prescribed drugs were characterized by their code in the anatomical therapeutic chemical classification system (ATC code). (iib) Causality assessment according to the Naranjo algorithm We used the Naranjo algorithm for causality assessment. All further relevant information, such as the duration of the AE, underlying diseases, clinical consequences (e.g. from hospital report), laboratory values, and patient-specific conditions were collected and used to determine the Naranjo score. The most likely associated drugs were the ones that reached the highest Naranjo score concerning the single analyzed AE. Naranjo distinguishes between definitive with a total score ≥9, probable with 5 < total score < 8, possible with 1 < total score < 4 and doubtful with a total score ≤ 0 [21, 22]. Inconclusive evaluations in all steps (i, ii.a, and ii.b) were discussed and finalized by mutual agreement in an expert panel. This panel consisted of four experienced clinical pharmacists. Statistical analysis To ensure comparable patient parameters between the three LTC facilities independent of the allocation to a single facility, main patient parameters were statistically analyzed. For this purpose, a Kruskal Wallis test with pairwise comparison was performed. Analyzed parameters were age, gender, number of diagnoses and number of continuous and on demand drugs, as well as the number of AEs in the patients and the maximum Naranjo score per patient. The data analysis was performed using IBM SPSS Statistics Version 25.0 (IBM Corporation, Armonk, NY, USA) and Microsoft Office Excel 2013 (Microsoft Corporation, Redmond, WA, USA). P-values ≤ 0.05 were considered as statistically significant. Results Patient characteristics In the participating parts of the LTC facilities, 182 patients were potentially available for the study and 154 met the inclusion criteria. From these, 104 patients or their legal guardian gave their informed consent as well as their responsible physician and were enrolled in the study. Patients were mostly female (72.1%) and in median 86 (range: 66–101) years old (Table 1). Patients did not differ between the three LTC facilities according to the following parameters: age (p = 0.311), gender (p = 0.684), number of diagnoses (p = 0.070) and number of continuous (p = 0.629) and on demand drugs (p = 0.911).Table 1 Characteristics of patients included in the study with frequency of documented diagnoses, main ATC classes and main active substances Characteristics Value Patients, total, n 104 Patients in facility of welfare ownership, n (%) 34 (32.7%) Patients in facility of municipal ownership, n (%) 30 (28.8%) Patients in facility of ownership by private association, n (%) 40 (38.5%) Female, n (%) 75 (72.1%) Length of residence (months), median (Q25/Q75; min–max) 31 (12/63; 1–414) Age (years), median (Q25/Q75; min–max) 86 (78/90; 66–101) Documented diagnoses, median (Q25/Q75; min–max) 15 (10/21; 3–35) No. of continuous drugs, median (Q25/Q75; min–max) 8 (6/10; 2–18) No. of on demand medication, median (Q25/Q75; min–max) 2 (1/3; 1–6) Documented diagnosisa Hypertension, n (%) 82 (78.8%) Dementia, n (%) 69 (66.3%) Diabetes, n (%) 41 (39.4%) Heart failure, n (%) 32 (30.8%) Atrial fibrillation, n (%) 32 (30.8%) Renal failure, n (%) 24 (23.1%) Osteoporosis, n (%) 19 (18.3%) Stroke, n (%) 17 (16.3%) Main ATC classesb C (cardiovascular system), n (%) 236 (28.7%) N (nervous system), n (%) 216 (26.3%) A (alimentary tract and metabolism), n (%) 164 (20.0%) B (blood and blood-forming organs), n (%) 68 (8.3%) H (systemic hormonal preparations, excluding sex hormones and insulins), n (%) 27 (3.3%) Main active substancesb Torasemide, n (%) 47 (5.7%) Pantoprazole, n (%) 40 (4.9%) Ramipril, n (%) 35 (4.3%) Acetylsalicylic acid, n (%) 33 (4.0%) Metoprolol, n (%) 23 (2.8%) ATC anatomical therapeutic chemical/defined daily dose classification, Q25/Q75 first and third quartile aOrder is based on the most relevant diagnoses found in literature data to geriatric patients bAccording to the documented continuous drugs (i) AE identification From a total of 104 patients, at least 1 AE was identified in 103 (99.0%). We identified 424 AEs, with a detected median of 4 (Q25/Q75: 2/5, range 1–14) AEs per patient, which equals 2.05 AEs per resident month. The identified AEs and the number of affected patients are shown in Table 2. The system organ classes renal and urinary disorder (87 patients), gastrointestinal disorder (43 patients), skin and subcutaneous tissue disorders (37 patients) were most common in our patient collective. Altogether, 72 different AE categories were detected, 185 AEs were identified in the patient records and 195 AEs by the nurses’ interviews, with 44 AEs in concordance of both methods. We found a significant difference in the detected number of AEs between the observed LTC facilities (p = 0.020). Following the pairwise comparison, we only found differences between the municipal LTC facility with 3 (Q25/Q75: 2/4) AEs and the private LTC facility with a median of 4 (Q25/Q75: 3/6.25) AEs (p = 0.022).Table 2 Identified adverse evnts (n = 424) according to CTCAE and affected patients (n = 104) System organ class Number of identified AEs, n (%) Affected patients, n (%) AE with number of affected patientsa (n) Renal and urinary disorders 88 (20.8) 87 (83.7) Urinary incontinence (87), urinary tract pain (1) Gastrointestinal disorders 55 (13.0) 43 (41.3) Constipation (22), vomiting (16), diarrhea (10), blackened stools (3), nausea (2), lower gastrointestinal bleeding (1), periodontal disease (1) Psychiatric disorders 55 (13.0) 35 (33.7) Confusion (21), restlessness (12), defensive behavior (8), insomnia (5), depression (4), anxiety (2), hallucinations (1), personality change (1), psychiatric disorders—other specify (1) Skin and subcutaneous tissue disorders 50 (11.8) 37 (35.6) Intertrigo (9), dry skin (7), hyperhidrosis (7), skin ulceration (6), local redness (5), pruritus (4), purpura (4), skin and subcutaneous tissue disorders—other specify (3), skin induration (1), urticaria (2), alopecia (1), angioedema (1) Metabolism and nutritional disorders 41 (9.7) 27 (26.0) Hyperglycemia (26), hypoglycemia (15) Musculoskeletal and connective tissue disorders 40 (9.4) 33 (31.7) Arthralgia (14), pain in extremity (12), back pain (6), arthritis (4), musculoskeletal and connective tissue disorders—other specify (3), general muscle weakness (1) Nervous system disorders 39 (9.2) 31 (29.8) Dizziness (11), somnolence (10), headache (4), syncope (3), ataxia (2), cognitive disturbance (2), paresthesia (2), depressed level of consciousness (1), lethargy (1), neuralgia (1), seizure (1), spasticity (1) Injury, poisoning and procedural complications 14 (3.3) 14 (13.5) Fall (14) Vascular disorders 11 (2.6) 11 (10.6) Hematoma (10), flushing (1) Infections and infestations 8 (1.9) 7 (6.7) Skin infection (4), vulval infection (2), conjunctivitis infective (1), stoma site infection (1) General disorders and administration site conditions 7 (1.7) 7 (6.7) Edema limbs (3), pain (3), fatigue (1) Respiratory, thoracic and mediastinal disorders 7 (1.7) 6 (5.8) Dyspnea (4), cough (1), epistaxis (1), respiratory, thoracic and mediastinal disorders—other specify (1) Ear and labyrinth disorders 3 (0.7) 3 (2.9) Hearing impaired (2), tinnitus (1) Cardiac disorders 2 (0.5) 2 (1.9) Chest pain—cardiac (1), palpitations (1) Eye disorders 2 (0.5) 1 (1.0) Blurred vision (1), glaucoma (1) Investigations 2 (0.5) 2 (1.9) Weight gain (1), weight loss (1) AE(s) adverse event(s), CTCAE common terminology criteria for adverse events aMultiple categories per patient possible (ii.a) Review of summary of product characteristics To analyze the concomitantly administered drugs, we assessed 3725 combinations of AEs and corresponding drugs. For this analysis five drug/AE pairs had to be excluded because no information from the SmPC was available (moisturizing eye drops, medical device). Considering every identified AE, patients had a median of 3 potentially causing drugs according to the SmPC, with a range from 0 to 11 drugs (Q25/Q75: 2/4; details in Fig. 1). The most frequently (n ≥ 10) detected AEs and the affected system organ classes are shown in Table 3. The ATC classes prescribed most often (C, N, A, B, H) were frequently among the potentially causing drugs for the most common system organ classes (Fig. 2).Fig. 1 Number of detected adverse events versus number of potentially causing drugs according Summary of Product Charactetistics. AE(s) adverse event(s), SmPC summary of products characteristics Table 3 Median number of potentially causing drugs according Summary of Product Charactersitics and corresponding Naranjo Score per patient (n = 104) for the most frequently detected (≥10) adverse events (AEs) and for their corresponding System organ classes (all 424 detected AE included) System organ class and ... most frequent AE Number (n) Median number of potentially causing drugs per patient [range] Median Naranjo score [range] Renal and urinary disorders 88 2 [0–5] 0 [−1–2] ... Urinary incontinence 87 2 [0–5] 0 [−1–2] Gastrointestinal disorders 55 5 [0–11] 2 [0–4] ... Constipation 22 5 [0–10] 0 [0–3] ... Vomiting 16 7.5 [2–10] 2 [0–4] ... Diarrhea 10 4.5 [2–11] 3 [0–3] Psychiatric disorders 55 3 [0–7] 0 [−1–9] ... Confusion 21 3 [1–7] 1 [0–8] ... Restlessness 12 3 [0–4] 0 [−1–3] Metabolism and nutrition disorders 41 3 [0–5] 1 [0–5] ... Hyperglycemia 26 3 [0–4] 1 [0–1] ... Hypoglycemia 15 3 [1–5] 4 [2–5] Musculoskeletal and connective tissue disorders 40 3 [0–8] 2 [−1–3] ... Arthralgia 14 3 [0–6] 1 [−1–3] ... Pain in extremity 12 2 [1–8] 2 [0–3] Nervous system disorders 39 4 [0–10] 1 [−1–7] ... Dizziness 11 5 [1–10] 2 [0–7] ... Somnolence 10 4 [3–7] 3 [0–5] Injury, poisoning and procedural complications 14 6 [3–11] 2 [0–3] ... Fall 14 6 [3–11] 2 [0–3] Vascular disorders 11 1 [0–3] 1 [0–3] ... Hematoma 10 1 [0–3] 0.5 [0–2] AE(s) Adverse Event(s), SmPC Summary of product characteristics Fig. 2 Potentially causing drugs according Summary of Product Characteristics differentiated into the ATC classes for the most frequently detected system organ classes. AE(s) adverse event(s), ATC anatomical therapeutic chemical/defined daily dose classification, CTCAE common criteria for the terminology of adverse events, SmPC summary of products characteristics. ATC classes: A: alimentary tract and metabolism, B: blood and blood forming organs, C: cardiovascular system, H: systemic hormonal preparations, excluding sex hormones and insulins, N: nervous system (ii.b) Causality assessment according to the Naranjo algorithm All 3730 drug/AE pairs were included in the causality assessment. From the 424 identified AEs, 198 (46.9%) were classified as ADR with “doubtful”, 218 (51.2%) “possible”, 7 (1.7%) “probable”, and 1 (0.2%) “definitive” cause (Table 4). We found no significant differences in the maximum Naranjo scores per patient between the three LTC facilities (p = 0.964). On the basis of 424 detected AEs, only 1 drug in 84 AEs (19.8%) and several drugs in 340 AEs (80.2%) reached the highest score (Table 4). According to Naranjo these need to be considered as the most likely causing drug(s).Table 4 Results of the adverse event drug causality assessment according to the Naranjo algorithm Naranjo Score per AE Identified AE [n] Number of affected patientsa, [n] Classification Identified AE per class, n (%) Number of AE with one/several highest Naranjo drug(s) [n] −1 22 17 Doubtful 199 (46.9) 38/161 0 177 92 1 68 48 Possible 217 (51.2) 38/179 2 90 51 3 41 30 4 18 18 5 3 3 Probable 7 (1.7) 7/0 6 2 2 7 1 1 8 1 1 9 1 1 Definitive 1 (0.2) 1/0 AE(s) adverse event(s) aSeveral AEs per patient possible The probable and definitive ADRs were as follows: angioedema (severity grade according to CTCAE 4) induced by enalapril, urticaria (grade 2) induced by amoxicillin and clavulanic acid, hypoglycemia (grade 1) induced by insulin glargine, paresthesia (grade 2) induced by tapentadol and a complex case of occurring hallucinations (grade 3) in combination with confusion (grade 3), dizziness (grade 3) and somnolence (grade 3, in total 4 detected AEs) which were attributable to digitoxin (highest Naranjo score). In this case, the patient also received high doses of oxycodone and duloxetine. It can be seen as a mixed intoxication based on the hospital report. In the algorithm, digitoxin reached a one-point higher score than oxycodone/duloxetine because measurement of the increased blood level was available only for digitoxin. All of the detected AEs and ADRs were managed adequately by the nurses, for example, by informing a physician or arranging a hospital admission for the affected patient. Thus, no further action was required due to this study. Discussion In our study we addressed AEs in geriatric patients living in LTC facilities. We assessed which type of AEs occurred and also investigated potential additive effects of polypharmacy. With nearly all (99%) patients affected by AEs, we demonstrated the relevance of this topic. We found the identified AEs potentially caused by up to 11 different administered drugs. The Naranjo algorithm showed at least possible drug causes in half of these AEs. Thereby, multiple drugs were equally likely involved 80% of the time. Our results point out that AEs should be systematically recorded in routine practice in LTC facilities. In order to prevent ADRs, additive effects need to be considered in any strategies developed. Prevalence of AE and ADR in LTC residents More than half of our identified AEs could be associated with drug use. Our rate of probable and definitive ADRs was similar to other studies in the LTC setting (0.04 vs. up to 0.10 ADRs per observed resident month), although studies should be compared with caution [9, 23]. Nevertheless, the causality assessment leaves us with a high number of possible ADRs. Especially for AEs which were ongoing for a longer period, causality assessment was challenging in the routine setting. Information to evaluate the exact temporal connection between AE and drug use was frequently missing and therefore could have led to lower Naranjo scores. To resolve this problem, a regular and structured routine assessment of AEs and potentially causing drugs might increase the chance to identify ADRs and protect patients from the consequences. Our overall rate of identified AEs was higher than results seen in other studies (2.05 AEs vs 0.03–0.12 per observed resident month) [9, 23]. This indicates that we identified a noticeable amount of the general symptom burden of LTC residents that results from underlying diseases or age-related changes. This is consistent with the fact that incontinence, pain, sleep disorders and psychopathological symptoms are widely found in LTC residents [24]. Therefore, a regular routine AE assessment can support ADR detection as well as structured symptom evaluation. Additive effects of polypharmacy The suspected AE was listed as an ADR in the respective SmPC in a median of 3 and up to 11 administered drugs per patient. In 80% of all identified AEs, various drugs reached the highest Naranjo score simultaneously. This means that they were equally likely to cause the AE. This coincidence can increase the chance of AE occurrence independently from single causality scores. This result also raises the question whether ADRs resulting from additive effects have been underestimated. In cases with “probable” or “definitive” ADRs (Naranjo ≥5), we found results from only one drug with the highest Naranjo score; however, in four of these AEs, the drug with the highest Naranjo (digitoxin) was only part of a mixed intoxication with duloxetine and oxycodone based on the hospital report for the affected patient. In this case, the sole consideration of the causality assessment could mask an additive effect of at least 3 concomitantly given drugs. This shows that additive effects need to be considered in every detected AE independently from the single causality. Besides ADRs from well-known drug classes (e.g. vascular ADRs from drugs affecting blood and blood-forming organs), in a substantial amount of AEs, we found involvement of varying ATC-classes that are less familiar (e.g. nervous system ADRs in drugs affecting the cardiovascular system). This underlines the complexity of geriatric patient treatment and the need for interdisciplinary medication reviews that include an assessment of drug-related problems, such as drug-drug interactions, potentially inappropriate medication, as well as ADRs [25, 26]. In routine care, however, potential additive effects are often not taken into account. In particular, new and unclear symptoms could be misinterpreted as new diseases and sometimes even lead to prescribing cascades [27, 28]. Implications for practice Firstly, our study shows the need for a good data base and a regular routine assessment of AEs that occur in LTC facilities. We found a very low concordance rate of only 10% between AEs detected in nurses’ interviews and those mentioned in the patient record analysis. This demonstrates the potential of information loss in LTC facilities due to heterogeneous and incomplete AE documentation [29]. It also indicates the potential of recall bias in the nurses. The identification of every occurring AE allows a better assessment of simultaneously occurring events. We found in our study, for example, a combination of vomiting and diarrhea that indicated an infection rather than an ADR. Furthermore, the information on patients’ current symptoms contributes to appropriate proposals for medication changes in cases of identified ADRs. Secondly, our results support the development of strategies with improved consideration of the additive effects of polypharmacy. Combining an AE assessment with structured medication reviews improves the drug cause analysis of AEs as well as the detection and interpretation of drug-related problems. Ongoing prospective evaluation of AEs and potential drug-related causes contributes to prevent patients from experiencing negative events. This process could be further accelerated by electronic assistance. Electronic documentation of AEs and computer-assisted signal detection of ADRs can support problem solving in a narrow timeframe since physicians and pharmacists are usually not permanently present in the LTC facilities [11, 30]. Database-supported comparison of the events with patients’ medication can assist pharmacists in a comprehensive medication review. Currently, such electronic solutions are rarely used in the LTC setting in Germany. They could also support future research by providing information on the additive effects of various combined drugs and underlying diseases. Thirdly, our data suggest the need for improvement in interdisciplinary communication in LTC facilities. In interprofessional teams with nurses, pharmacists and physicians, systematic information about AEs, medication reviews and actual health conditions could be transmitted more effectively in patient-orientated practice. Conclusion Nearly every long-term care resident suffered from adverse events (AEs), with half of them at least possibly caused by drugs. In four fifths of these AEs, several concomitantly given drugs were equally associated causes. Therefore, potential additive effects need to be considered independently from single causality and should be more focused in further research. A routinely implemented structured search for AEs and additive effects of polypharmacy contributes to medication reviews and interdisciplinary collaboration and will help to meet the needs of this complex patient collective and to protect them from negative consequences. Acknowledgements We thank all participating co-workers of the LTC facilities and the attending physicians for their support. Furthermore, we thank PhD Johanna Freyer for her assistance with the ethics approval and Katharine Worthington for language editing. Funding A co-worker of the study (Monika Lexow) was financially supported in part by the Lesmueller Foundation, Munich, Germany, the German Pharmacist Foundation, Berlin, Germany and the Pharmacist Foundation Westfalen-Lippe, Münster, Germany. Author Contribution Conceptualization: Monika Lexow, Kathrin Wernecke, Ralf Sulzer, Thilo Bertsche, Susanne Schiek; methodology: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; formal analysis and investigation: Kathrin Wernecke, Susanne Schiek; investigation: Monika Lexow, Kathrin Wernecke; writing, original draft preparation: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; writing, review and editing: Monika Lexow, Kathrin Wernecke, Ralf Sultzer, Gordian L. Schmid, Thilo Bertsche, Susanne Schiek; visualization: Monika Lexow, Kathrin Wernecke, Susanne Schiek; supervision: Thilo Bertsche, Susanne Schiek; project administration: Monika Lexow; funding acquisition: Thilo Bertsche, Susanne Schiek. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with ethical guidelines Conflict of interest M. Lexow, K. Wernecke, G.L. Schmid, R. Sultzer, T. Bertsche, and S. Schiek declare that they have no competing interests. Ethical standards All procedures performed in this study involving human participants were approved both by the ethics committee of the Faculty of Medicine of Leipzig University as well as the ethics committee of the State Chamber of Physicians of Saxony (reference: 231/13-ff and EK-allg-26/14-1) and have been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All participants gave their informed consent prior to inclusion in the study. The authors M. Lexow and K. Wernecke contributed equally to the manuscript. The authors T. Bertsche and S. Schiek contributed equally to the manuscript. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Recovering
ReactionOutcome
CC BY
33090261
20,027,693
2021-08
What was the outcome of reaction 'Dizziness'?
Considering additive effects of polypharmacy : Analysis of adverse events in geriatric patients in long-term care facilities. BACKGROUND Potential additive effects of polypharmacy are rarely considered in adverse events of geriatric patients living in long-term care facilities. Our aim, therefore, was to identify adverse events in this setting and to assess plausible concomitant drug causes. METHODS A cross-sectional observational study was performed in three facilities as follows: (i) adverse event identification: we structurally identified adverse events using nurses' interviews and chart review. (ii) Analysis of the concomitantly administered drugs per patient was performed in two ways: (ii.a) a review of summary of product characteristics for listed adverse drug reactions to identify possible causing drugs and (ii.b) a causality assessment according to Naranjo algorithm. RESULTS (i) We found 424 adverse events with a median of 4 per patient (range 1-14) in 103 of the 104 enrolled patients (99%). (ii.a) We identified a median of 3 drugs (range 0-11) with actually occurring adverse events listed as an adverse drug reaction in the summary of product characteristics. (ii.b) Causality was classified in 198 (46.9%) of adverse events as "doubtful," in 218 (51.2%) as "possible," in 7 (1.7%) as "probable," and in 1 (0.2%) adverse event as a "definitive" cause of the administered drugs. In 340 (80.2%) of all identified adverse events several drugs simultaneously reached the highest respective Naranjo score. CONCLUSIONS Patients in long-term facilities frequently suffer from many adverse events. Concomitantly administered drugs have to be frequently considered as plausible causes for adverse events. These additive effects of drugs should be more focused in patient care and research. Introduction Geriatric patients in long-term care (LTC) facilities are multimorbid and, therefore, suffer from many (non)specific symptoms and geriatric syndromes [1]. Disease-related symptoms should be distinguished from adverse drug reactions (ADR) that result from drug therapy [2]. The latter can lead to hospital admissions and have a considerable impact on morbidity and mortality with high costs in the health care system [3–5]. Polypharmacy makes a significant contribution to the clinical consequences deriving from ADRs in geriatric patients [6]. For this reason, a structured analysis of adverse events (AE) and drug-related causes in these patients is of high interest for routine care. Distinguishing whether an observed AE was caused by a disease (i.e. symptom) or by a drug (ADR) poses a challenge for healthcare professionals [7, 8]. The correct attribution is required for appropriate treatment strategies but can result only from structured detection, analysis and classification. Geriatric patients are frequently cognitively impaired or suffer from speech or hearing disorders. Hence, information provided by the patients is often insufficient. In LTC facilities, therefore, chart documentation and nurses’ interviews are the most valuable sources for AE detection [9, 10]. So far, no specific method exists to analyze and classify AE in LTC facility residents with polypharmacy. The Naranjo algorithm has previously been used for causality assessment in this collective [11, 12]. It allows a detailed assessment of every detected AE and every single administered drug. This algorithm provides further information on drug-related causes when combined with established methods for patient safety, such as drug-drug interactions and potentially inappropriate medications [13]. Causality scores like the Naranjo score, however, do not consider simultaneously contributing drugs. For some ADRs, it has been shown that the number of specific drugs causes their clinical manifestations. For example, patients are exposed to an increased risk of falling when they take two or more drugs which increase the risk of falling [14]. Concerning anticholinergic ADRs, it is common to calculate an anticholinergic burden to quantify the risk for an adverse outcome [15]. Little is known, however, about additive drug effects in other events. Therefore, data about potential additive effects in this vulnerable patient collective are of great interest for routine care. The aim of this study was to identify AEs occurring in LTC facility patients and to assess plausible concomitant drug causes. Patients, material and methods Definitions We defined an AE as an outcome that occurs while a patient is taking a drug, but is not or not necessarily attributable to it and an ADR as an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product [16]. We used the term drug not only for the effective substance but for the whole product prescribed in the medication chart of the patient. A drug therefore could contain more than one active substance. We considered all drugs administered to the patient during the acquisition period. Continuous and on-demand medications were assessed separately because the temporal relationship between AE and administration of the drug could be different in that case. Participants and setting We conducted a cross-sectional observation study in three LTC facilities in Germany. After written informed consent of the residents or their legal representative and the responsible general practitioner, residents in the participating LTC facilities were enrolled in the study. We included residents of facilities with different ownerships (welfare, municipal or private associations) to approach a representative sample of 100 residents. Inclusion criteria were: informed consent, age ≥65 years, long-term/chronic medicines ≥3 and multimorbidity with ≥3 comorbidities at the time of recruitment, more than 8 weeks stay in the LTC facility, and a life expectancy of more than 6 months according to nurses’ present information. The study was conducted over a time period of 10 months. Study design and data collection We conducted a structured analysis of AEs. (i) AE identification We used two complement sources of information for our structured data collection: Firstly, an interview about individual AEs with nurses involved in daily care and, secondly, a review of residents’ records (electronic and chart documentation, laboratory values) for documented events and their temporal occurrence. To ensure standardized identification of AEs, a checklist of events was applied to both methods. The listed events comprised the most relevant AEs or ADRs for geriatric patients and LTC residents based on the literature [17–19]: blackened stool, bleeding/hematoma, confusion/disorientation, constipation, depression/anxiety, diarrhea, dizziness/vertigo, dry mouth, ear disorders, eye disorders, falls, hallucination, hyperglycemia/hypoglycemia, hyperhidrosis, hyperkalemia/hypokalemia, hypernatremia/hyponatremia, nausea, pain, restlessness, skin disorders/pruritus, insomnia, urinary incontinence, vomiting (in alphabetical order). Additional relevant reported or documented events were collected as well. We considered reported and documented symptoms during a time period of the prior 30 days (considered as 1 resident month) for new and continuous symptoms. Data collection was performed at two measurement points per patient at intervals of 6–8 weeks by two clinical pharmacists. All detected AEs and the corresponding system organ class were classified based on the common terminology criteria for adverse events (CTCAE) [20]. (iia) Review of summary of product characteristics We systematically collected data from the medical charts (continuous drugs, on demand drugs and their frequency of use, date of first administration). We checked all summaries of product characteristics (SmPCs) of the actually administered drugs for listed ADRs. A drug would be considered as “potentially causing” if the listed ADR in the SmPC represented a synonym for the detected AE or possibly caused it (e.g. dizziness in cases of falls). For the analysis of additive effects, we counted the number of potentially causing drugs. Prescribed drugs were characterized by their code in the anatomical therapeutic chemical classification system (ATC code). (iib) Causality assessment according to the Naranjo algorithm We used the Naranjo algorithm for causality assessment. All further relevant information, such as the duration of the AE, underlying diseases, clinical consequences (e.g. from hospital report), laboratory values, and patient-specific conditions were collected and used to determine the Naranjo score. The most likely associated drugs were the ones that reached the highest Naranjo score concerning the single analyzed AE. Naranjo distinguishes between definitive with a total score ≥9, probable with 5 < total score < 8, possible with 1 < total score < 4 and doubtful with a total score ≤ 0 [21, 22]. Inconclusive evaluations in all steps (i, ii.a, and ii.b) were discussed and finalized by mutual agreement in an expert panel. This panel consisted of four experienced clinical pharmacists. Statistical analysis To ensure comparable patient parameters between the three LTC facilities independent of the allocation to a single facility, main patient parameters were statistically analyzed. For this purpose, a Kruskal Wallis test with pairwise comparison was performed. Analyzed parameters were age, gender, number of diagnoses and number of continuous and on demand drugs, as well as the number of AEs in the patients and the maximum Naranjo score per patient. The data analysis was performed using IBM SPSS Statistics Version 25.0 (IBM Corporation, Armonk, NY, USA) and Microsoft Office Excel 2013 (Microsoft Corporation, Redmond, WA, USA). P-values ≤ 0.05 were considered as statistically significant. Results Patient characteristics In the participating parts of the LTC facilities, 182 patients were potentially available for the study and 154 met the inclusion criteria. From these, 104 patients or their legal guardian gave their informed consent as well as their responsible physician and were enrolled in the study. Patients were mostly female (72.1%) and in median 86 (range: 66–101) years old (Table 1). Patients did not differ between the three LTC facilities according to the following parameters: age (p = 0.311), gender (p = 0.684), number of diagnoses (p = 0.070) and number of continuous (p = 0.629) and on demand drugs (p = 0.911).Table 1 Characteristics of patients included in the study with frequency of documented diagnoses, main ATC classes and main active substances Characteristics Value Patients, total, n 104 Patients in facility of welfare ownership, n (%) 34 (32.7%) Patients in facility of municipal ownership, n (%) 30 (28.8%) Patients in facility of ownership by private association, n (%) 40 (38.5%) Female, n (%) 75 (72.1%) Length of residence (months), median (Q25/Q75; min–max) 31 (12/63; 1–414) Age (years), median (Q25/Q75; min–max) 86 (78/90; 66–101) Documented diagnoses, median (Q25/Q75; min–max) 15 (10/21; 3–35) No. of continuous drugs, median (Q25/Q75; min–max) 8 (6/10; 2–18) No. of on demand medication, median (Q25/Q75; min–max) 2 (1/3; 1–6) Documented diagnosisa Hypertension, n (%) 82 (78.8%) Dementia, n (%) 69 (66.3%) Diabetes, n (%) 41 (39.4%) Heart failure, n (%) 32 (30.8%) Atrial fibrillation, n (%) 32 (30.8%) Renal failure, n (%) 24 (23.1%) Osteoporosis, n (%) 19 (18.3%) Stroke, n (%) 17 (16.3%) Main ATC classesb C (cardiovascular system), n (%) 236 (28.7%) N (nervous system), n (%) 216 (26.3%) A (alimentary tract and metabolism), n (%) 164 (20.0%) B (blood and blood-forming organs), n (%) 68 (8.3%) H (systemic hormonal preparations, excluding sex hormones and insulins), n (%) 27 (3.3%) Main active substancesb Torasemide, n (%) 47 (5.7%) Pantoprazole, n (%) 40 (4.9%) Ramipril, n (%) 35 (4.3%) Acetylsalicylic acid, n (%) 33 (4.0%) Metoprolol, n (%) 23 (2.8%) ATC anatomical therapeutic chemical/defined daily dose classification, Q25/Q75 first and third quartile aOrder is based on the most relevant diagnoses found in literature data to geriatric patients bAccording to the documented continuous drugs (i) AE identification From a total of 104 patients, at least 1 AE was identified in 103 (99.0%). We identified 424 AEs, with a detected median of 4 (Q25/Q75: 2/5, range 1–14) AEs per patient, which equals 2.05 AEs per resident month. The identified AEs and the number of affected patients are shown in Table 2. The system organ classes renal and urinary disorder (87 patients), gastrointestinal disorder (43 patients), skin and subcutaneous tissue disorders (37 patients) were most common in our patient collective. Altogether, 72 different AE categories were detected, 185 AEs were identified in the patient records and 195 AEs by the nurses’ interviews, with 44 AEs in concordance of both methods. We found a significant difference in the detected number of AEs between the observed LTC facilities (p = 0.020). Following the pairwise comparison, we only found differences between the municipal LTC facility with 3 (Q25/Q75: 2/4) AEs and the private LTC facility with a median of 4 (Q25/Q75: 3/6.25) AEs (p = 0.022).Table 2 Identified adverse evnts (n = 424) according to CTCAE and affected patients (n = 104) System organ class Number of identified AEs, n (%) Affected patients, n (%) AE with number of affected patientsa (n) Renal and urinary disorders 88 (20.8) 87 (83.7) Urinary incontinence (87), urinary tract pain (1) Gastrointestinal disorders 55 (13.0) 43 (41.3) Constipation (22), vomiting (16), diarrhea (10), blackened stools (3), nausea (2), lower gastrointestinal bleeding (1), periodontal disease (1) Psychiatric disorders 55 (13.0) 35 (33.7) Confusion (21), restlessness (12), defensive behavior (8), insomnia (5), depression (4), anxiety (2), hallucinations (1), personality change (1), psychiatric disorders—other specify (1) Skin and subcutaneous tissue disorders 50 (11.8) 37 (35.6) Intertrigo (9), dry skin (7), hyperhidrosis (7), skin ulceration (6), local redness (5), pruritus (4), purpura (4), skin and subcutaneous tissue disorders—other specify (3), skin induration (1), urticaria (2), alopecia (1), angioedema (1) Metabolism and nutritional disorders 41 (9.7) 27 (26.0) Hyperglycemia (26), hypoglycemia (15) Musculoskeletal and connective tissue disorders 40 (9.4) 33 (31.7) Arthralgia (14), pain in extremity (12), back pain (6), arthritis (4), musculoskeletal and connective tissue disorders—other specify (3), general muscle weakness (1) Nervous system disorders 39 (9.2) 31 (29.8) Dizziness (11), somnolence (10), headache (4), syncope (3), ataxia (2), cognitive disturbance (2), paresthesia (2), depressed level of consciousness (1), lethargy (1), neuralgia (1), seizure (1), spasticity (1) Injury, poisoning and procedural complications 14 (3.3) 14 (13.5) Fall (14) Vascular disorders 11 (2.6) 11 (10.6) Hematoma (10), flushing (1) Infections and infestations 8 (1.9) 7 (6.7) Skin infection (4), vulval infection (2), conjunctivitis infective (1), stoma site infection (1) General disorders and administration site conditions 7 (1.7) 7 (6.7) Edema limbs (3), pain (3), fatigue (1) Respiratory, thoracic and mediastinal disorders 7 (1.7) 6 (5.8) Dyspnea (4), cough (1), epistaxis (1), respiratory, thoracic and mediastinal disorders—other specify (1) Ear and labyrinth disorders 3 (0.7) 3 (2.9) Hearing impaired (2), tinnitus (1) Cardiac disorders 2 (0.5) 2 (1.9) Chest pain—cardiac (1), palpitations (1) Eye disorders 2 (0.5) 1 (1.0) Blurred vision (1), glaucoma (1) Investigations 2 (0.5) 2 (1.9) Weight gain (1), weight loss (1) AE(s) adverse event(s), CTCAE common terminology criteria for adverse events aMultiple categories per patient possible (ii.a) Review of summary of product characteristics To analyze the concomitantly administered drugs, we assessed 3725 combinations of AEs and corresponding drugs. For this analysis five drug/AE pairs had to be excluded because no information from the SmPC was available (moisturizing eye drops, medical device). Considering every identified AE, patients had a median of 3 potentially causing drugs according to the SmPC, with a range from 0 to 11 drugs (Q25/Q75: 2/4; details in Fig. 1). The most frequently (n ≥ 10) detected AEs and the affected system organ classes are shown in Table 3. The ATC classes prescribed most often (C, N, A, B, H) were frequently among the potentially causing drugs for the most common system organ classes (Fig. 2).Fig. 1 Number of detected adverse events versus number of potentially causing drugs according Summary of Product Charactetistics. AE(s) adverse event(s), SmPC summary of products characteristics Table 3 Median number of potentially causing drugs according Summary of Product Charactersitics and corresponding Naranjo Score per patient (n = 104) for the most frequently detected (≥10) adverse events (AEs) and for their corresponding System organ classes (all 424 detected AE included) System organ class and ... most frequent AE Number (n) Median number of potentially causing drugs per patient [range] Median Naranjo score [range] Renal and urinary disorders 88 2 [0–5] 0 [−1–2] ... Urinary incontinence 87 2 [0–5] 0 [−1–2] Gastrointestinal disorders 55 5 [0–11] 2 [0–4] ... Constipation 22 5 [0–10] 0 [0–3] ... Vomiting 16 7.5 [2–10] 2 [0–4] ... Diarrhea 10 4.5 [2–11] 3 [0–3] Psychiatric disorders 55 3 [0–7] 0 [−1–9] ... Confusion 21 3 [1–7] 1 [0–8] ... Restlessness 12 3 [0–4] 0 [−1–3] Metabolism and nutrition disorders 41 3 [0–5] 1 [0–5] ... Hyperglycemia 26 3 [0–4] 1 [0–1] ... Hypoglycemia 15 3 [1–5] 4 [2–5] Musculoskeletal and connective tissue disorders 40 3 [0–8] 2 [−1–3] ... Arthralgia 14 3 [0–6] 1 [−1–3] ... Pain in extremity 12 2 [1–8] 2 [0–3] Nervous system disorders 39 4 [0–10] 1 [−1–7] ... Dizziness 11 5 [1–10] 2 [0–7] ... Somnolence 10 4 [3–7] 3 [0–5] Injury, poisoning and procedural complications 14 6 [3–11] 2 [0–3] ... Fall 14 6 [3–11] 2 [0–3] Vascular disorders 11 1 [0–3] 1 [0–3] ... Hematoma 10 1 [0–3] 0.5 [0–2] AE(s) Adverse Event(s), SmPC Summary of product characteristics Fig. 2 Potentially causing drugs according Summary of Product Characteristics differentiated into the ATC classes for the most frequently detected system organ classes. AE(s) adverse event(s), ATC anatomical therapeutic chemical/defined daily dose classification, CTCAE common criteria for the terminology of adverse events, SmPC summary of products characteristics. ATC classes: A: alimentary tract and metabolism, B: blood and blood forming organs, C: cardiovascular system, H: systemic hormonal preparations, excluding sex hormones and insulins, N: nervous system (ii.b) Causality assessment according to the Naranjo algorithm All 3730 drug/AE pairs were included in the causality assessment. From the 424 identified AEs, 198 (46.9%) were classified as ADR with “doubtful”, 218 (51.2%) “possible”, 7 (1.7%) “probable”, and 1 (0.2%) “definitive” cause (Table 4). We found no significant differences in the maximum Naranjo scores per patient between the three LTC facilities (p = 0.964). On the basis of 424 detected AEs, only 1 drug in 84 AEs (19.8%) and several drugs in 340 AEs (80.2%) reached the highest score (Table 4). According to Naranjo these need to be considered as the most likely causing drug(s).Table 4 Results of the adverse event drug causality assessment according to the Naranjo algorithm Naranjo Score per AE Identified AE [n] Number of affected patientsa, [n] Classification Identified AE per class, n (%) Number of AE with one/several highest Naranjo drug(s) [n] −1 22 17 Doubtful 199 (46.9) 38/161 0 177 92 1 68 48 Possible 217 (51.2) 38/179 2 90 51 3 41 30 4 18 18 5 3 3 Probable 7 (1.7) 7/0 6 2 2 7 1 1 8 1 1 9 1 1 Definitive 1 (0.2) 1/0 AE(s) adverse event(s) aSeveral AEs per patient possible The probable and definitive ADRs were as follows: angioedema (severity grade according to CTCAE 4) induced by enalapril, urticaria (grade 2) induced by amoxicillin and clavulanic acid, hypoglycemia (grade 1) induced by insulin glargine, paresthesia (grade 2) induced by tapentadol and a complex case of occurring hallucinations (grade 3) in combination with confusion (grade 3), dizziness (grade 3) and somnolence (grade 3, in total 4 detected AEs) which were attributable to digitoxin (highest Naranjo score). In this case, the patient also received high doses of oxycodone and duloxetine. It can be seen as a mixed intoxication based on the hospital report. In the algorithm, digitoxin reached a one-point higher score than oxycodone/duloxetine because measurement of the increased blood level was available only for digitoxin. All of the detected AEs and ADRs were managed adequately by the nurses, for example, by informing a physician or arranging a hospital admission for the affected patient. Thus, no further action was required due to this study. Discussion In our study we addressed AEs in geriatric patients living in LTC facilities. We assessed which type of AEs occurred and also investigated potential additive effects of polypharmacy. With nearly all (99%) patients affected by AEs, we demonstrated the relevance of this topic. We found the identified AEs potentially caused by up to 11 different administered drugs. The Naranjo algorithm showed at least possible drug causes in half of these AEs. Thereby, multiple drugs were equally likely involved 80% of the time. Our results point out that AEs should be systematically recorded in routine practice in LTC facilities. In order to prevent ADRs, additive effects need to be considered in any strategies developed. Prevalence of AE and ADR in LTC residents More than half of our identified AEs could be associated with drug use. Our rate of probable and definitive ADRs was similar to other studies in the LTC setting (0.04 vs. up to 0.10 ADRs per observed resident month), although studies should be compared with caution [9, 23]. Nevertheless, the causality assessment leaves us with a high number of possible ADRs. Especially for AEs which were ongoing for a longer period, causality assessment was challenging in the routine setting. Information to evaluate the exact temporal connection between AE and drug use was frequently missing and therefore could have led to lower Naranjo scores. To resolve this problem, a regular and structured routine assessment of AEs and potentially causing drugs might increase the chance to identify ADRs and protect patients from the consequences. Our overall rate of identified AEs was higher than results seen in other studies (2.05 AEs vs 0.03–0.12 per observed resident month) [9, 23]. This indicates that we identified a noticeable amount of the general symptom burden of LTC residents that results from underlying diseases or age-related changes. This is consistent with the fact that incontinence, pain, sleep disorders and psychopathological symptoms are widely found in LTC residents [24]. Therefore, a regular routine AE assessment can support ADR detection as well as structured symptom evaluation. Additive effects of polypharmacy The suspected AE was listed as an ADR in the respective SmPC in a median of 3 and up to 11 administered drugs per patient. In 80% of all identified AEs, various drugs reached the highest Naranjo score simultaneously. This means that they were equally likely to cause the AE. This coincidence can increase the chance of AE occurrence independently from single causality scores. This result also raises the question whether ADRs resulting from additive effects have been underestimated. In cases with “probable” or “definitive” ADRs (Naranjo ≥5), we found results from only one drug with the highest Naranjo score; however, in four of these AEs, the drug with the highest Naranjo (digitoxin) was only part of a mixed intoxication with duloxetine and oxycodone based on the hospital report for the affected patient. In this case, the sole consideration of the causality assessment could mask an additive effect of at least 3 concomitantly given drugs. This shows that additive effects need to be considered in every detected AE independently from the single causality. Besides ADRs from well-known drug classes (e.g. vascular ADRs from drugs affecting blood and blood-forming organs), in a substantial amount of AEs, we found involvement of varying ATC-classes that are less familiar (e.g. nervous system ADRs in drugs affecting the cardiovascular system). This underlines the complexity of geriatric patient treatment and the need for interdisciplinary medication reviews that include an assessment of drug-related problems, such as drug-drug interactions, potentially inappropriate medication, as well as ADRs [25, 26]. In routine care, however, potential additive effects are often not taken into account. In particular, new and unclear symptoms could be misinterpreted as new diseases and sometimes even lead to prescribing cascades [27, 28]. Implications for practice Firstly, our study shows the need for a good data base and a regular routine assessment of AEs that occur in LTC facilities. We found a very low concordance rate of only 10% between AEs detected in nurses’ interviews and those mentioned in the patient record analysis. This demonstrates the potential of information loss in LTC facilities due to heterogeneous and incomplete AE documentation [29]. It also indicates the potential of recall bias in the nurses. The identification of every occurring AE allows a better assessment of simultaneously occurring events. We found in our study, for example, a combination of vomiting and diarrhea that indicated an infection rather than an ADR. Furthermore, the information on patients’ current symptoms contributes to appropriate proposals for medication changes in cases of identified ADRs. Secondly, our results support the development of strategies with improved consideration of the additive effects of polypharmacy. Combining an AE assessment with structured medication reviews improves the drug cause analysis of AEs as well as the detection and interpretation of drug-related problems. Ongoing prospective evaluation of AEs and potential drug-related causes contributes to prevent patients from experiencing negative events. This process could be further accelerated by electronic assistance. Electronic documentation of AEs and computer-assisted signal detection of ADRs can support problem solving in a narrow timeframe since physicians and pharmacists are usually not permanently present in the LTC facilities [11, 30]. Database-supported comparison of the events with patients’ medication can assist pharmacists in a comprehensive medication review. Currently, such electronic solutions are rarely used in the LTC setting in Germany. They could also support future research by providing information on the additive effects of various combined drugs and underlying diseases. Thirdly, our data suggest the need for improvement in interdisciplinary communication in LTC facilities. In interprofessional teams with nurses, pharmacists and physicians, systematic information about AEs, medication reviews and actual health conditions could be transmitted more effectively in patient-orientated practice. Conclusion Nearly every long-term care resident suffered from adverse events (AEs), with half of them at least possibly caused by drugs. In four fifths of these AEs, several concomitantly given drugs were equally associated causes. Therefore, potential additive effects need to be considered independently from single causality and should be more focused in further research. A routinely implemented structured search for AEs and additive effects of polypharmacy contributes to medication reviews and interdisciplinary collaboration and will help to meet the needs of this complex patient collective and to protect them from negative consequences. Acknowledgements We thank all participating co-workers of the LTC facilities and the attending physicians for their support. Furthermore, we thank PhD Johanna Freyer for her assistance with the ethics approval and Katharine Worthington for language editing. Funding A co-worker of the study (Monika Lexow) was financially supported in part by the Lesmueller Foundation, Munich, Germany, the German Pharmacist Foundation, Berlin, Germany and the Pharmacist Foundation Westfalen-Lippe, Münster, Germany. Author Contribution Conceptualization: Monika Lexow, Kathrin Wernecke, Ralf Sulzer, Thilo Bertsche, Susanne Schiek; methodology: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; formal analysis and investigation: Kathrin Wernecke, Susanne Schiek; investigation: Monika Lexow, Kathrin Wernecke; writing, original draft preparation: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; writing, review and editing: Monika Lexow, Kathrin Wernecke, Ralf Sultzer, Gordian L. Schmid, Thilo Bertsche, Susanne Schiek; visualization: Monika Lexow, Kathrin Wernecke, Susanne Schiek; supervision: Thilo Bertsche, Susanne Schiek; project administration: Monika Lexow; funding acquisition: Thilo Bertsche, Susanne Schiek. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with ethical guidelines Conflict of interest M. Lexow, K. Wernecke, G.L. Schmid, R. Sultzer, T. Bertsche, and S. Schiek declare that they have no competing interests. Ethical standards All procedures performed in this study involving human participants were approved both by the ethics committee of the Faculty of Medicine of Leipzig University as well as the ethics committee of the State Chamber of Physicians of Saxony (reference: 231/13-ff and EK-allg-26/14-1) and have been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All participants gave their informed consent prior to inclusion in the study. The authors M. Lexow and K. Wernecke contributed equally to the manuscript. The authors T. Bertsche and S. Schiek contributed equally to the manuscript. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Recovering
ReactionOutcome
CC BY
33090261
20,027,693
2021-08
What was the outcome of reaction 'Hallucination'?
Considering additive effects of polypharmacy : Analysis of adverse events in geriatric patients in long-term care facilities. BACKGROUND Potential additive effects of polypharmacy are rarely considered in adverse events of geriatric patients living in long-term care facilities. Our aim, therefore, was to identify adverse events in this setting and to assess plausible concomitant drug causes. METHODS A cross-sectional observational study was performed in three facilities as follows: (i) adverse event identification: we structurally identified adverse events using nurses' interviews and chart review. (ii) Analysis of the concomitantly administered drugs per patient was performed in two ways: (ii.a) a review of summary of product characteristics for listed adverse drug reactions to identify possible causing drugs and (ii.b) a causality assessment according to Naranjo algorithm. RESULTS (i) We found 424 adverse events with a median of 4 per patient (range 1-14) in 103 of the 104 enrolled patients (99%). (ii.a) We identified a median of 3 drugs (range 0-11) with actually occurring adverse events listed as an adverse drug reaction in the summary of product characteristics. (ii.b) Causality was classified in 198 (46.9%) of adverse events as "doubtful," in 218 (51.2%) as "possible," in 7 (1.7%) as "probable," and in 1 (0.2%) adverse event as a "definitive" cause of the administered drugs. In 340 (80.2%) of all identified adverse events several drugs simultaneously reached the highest respective Naranjo score. CONCLUSIONS Patients in long-term facilities frequently suffer from many adverse events. Concomitantly administered drugs have to be frequently considered as plausible causes for adverse events. These additive effects of drugs should be more focused in patient care and research. Introduction Geriatric patients in long-term care (LTC) facilities are multimorbid and, therefore, suffer from many (non)specific symptoms and geriatric syndromes [1]. Disease-related symptoms should be distinguished from adverse drug reactions (ADR) that result from drug therapy [2]. The latter can lead to hospital admissions and have a considerable impact on morbidity and mortality with high costs in the health care system [3–5]. Polypharmacy makes a significant contribution to the clinical consequences deriving from ADRs in geriatric patients [6]. For this reason, a structured analysis of adverse events (AE) and drug-related causes in these patients is of high interest for routine care. Distinguishing whether an observed AE was caused by a disease (i.e. symptom) or by a drug (ADR) poses a challenge for healthcare professionals [7, 8]. The correct attribution is required for appropriate treatment strategies but can result only from structured detection, analysis and classification. Geriatric patients are frequently cognitively impaired or suffer from speech or hearing disorders. Hence, information provided by the patients is often insufficient. In LTC facilities, therefore, chart documentation and nurses’ interviews are the most valuable sources for AE detection [9, 10]. So far, no specific method exists to analyze and classify AE in LTC facility residents with polypharmacy. The Naranjo algorithm has previously been used for causality assessment in this collective [11, 12]. It allows a detailed assessment of every detected AE and every single administered drug. This algorithm provides further information on drug-related causes when combined with established methods for patient safety, such as drug-drug interactions and potentially inappropriate medications [13]. Causality scores like the Naranjo score, however, do not consider simultaneously contributing drugs. For some ADRs, it has been shown that the number of specific drugs causes their clinical manifestations. For example, patients are exposed to an increased risk of falling when they take two or more drugs which increase the risk of falling [14]. Concerning anticholinergic ADRs, it is common to calculate an anticholinergic burden to quantify the risk for an adverse outcome [15]. Little is known, however, about additive drug effects in other events. Therefore, data about potential additive effects in this vulnerable patient collective are of great interest for routine care. The aim of this study was to identify AEs occurring in LTC facility patients and to assess plausible concomitant drug causes. Patients, material and methods Definitions We defined an AE as an outcome that occurs while a patient is taking a drug, but is not or not necessarily attributable to it and an ADR as an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product [16]. We used the term drug not only for the effective substance but for the whole product prescribed in the medication chart of the patient. A drug therefore could contain more than one active substance. We considered all drugs administered to the patient during the acquisition period. Continuous and on-demand medications were assessed separately because the temporal relationship between AE and administration of the drug could be different in that case. Participants and setting We conducted a cross-sectional observation study in three LTC facilities in Germany. After written informed consent of the residents or their legal representative and the responsible general practitioner, residents in the participating LTC facilities were enrolled in the study. We included residents of facilities with different ownerships (welfare, municipal or private associations) to approach a representative sample of 100 residents. Inclusion criteria were: informed consent, age ≥65 years, long-term/chronic medicines ≥3 and multimorbidity with ≥3 comorbidities at the time of recruitment, more than 8 weeks stay in the LTC facility, and a life expectancy of more than 6 months according to nurses’ present information. The study was conducted over a time period of 10 months. Study design and data collection We conducted a structured analysis of AEs. (i) AE identification We used two complement sources of information for our structured data collection: Firstly, an interview about individual AEs with nurses involved in daily care and, secondly, a review of residents’ records (electronic and chart documentation, laboratory values) for documented events and their temporal occurrence. To ensure standardized identification of AEs, a checklist of events was applied to both methods. The listed events comprised the most relevant AEs or ADRs for geriatric patients and LTC residents based on the literature [17–19]: blackened stool, bleeding/hematoma, confusion/disorientation, constipation, depression/anxiety, diarrhea, dizziness/vertigo, dry mouth, ear disorders, eye disorders, falls, hallucination, hyperglycemia/hypoglycemia, hyperhidrosis, hyperkalemia/hypokalemia, hypernatremia/hyponatremia, nausea, pain, restlessness, skin disorders/pruritus, insomnia, urinary incontinence, vomiting (in alphabetical order). Additional relevant reported or documented events were collected as well. We considered reported and documented symptoms during a time period of the prior 30 days (considered as 1 resident month) for new and continuous symptoms. Data collection was performed at two measurement points per patient at intervals of 6–8 weeks by two clinical pharmacists. All detected AEs and the corresponding system organ class were classified based on the common terminology criteria for adverse events (CTCAE) [20]. (iia) Review of summary of product characteristics We systematically collected data from the medical charts (continuous drugs, on demand drugs and their frequency of use, date of first administration). We checked all summaries of product characteristics (SmPCs) of the actually administered drugs for listed ADRs. A drug would be considered as “potentially causing” if the listed ADR in the SmPC represented a synonym for the detected AE or possibly caused it (e.g. dizziness in cases of falls). For the analysis of additive effects, we counted the number of potentially causing drugs. Prescribed drugs were characterized by their code in the anatomical therapeutic chemical classification system (ATC code). (iib) Causality assessment according to the Naranjo algorithm We used the Naranjo algorithm for causality assessment. All further relevant information, such as the duration of the AE, underlying diseases, clinical consequences (e.g. from hospital report), laboratory values, and patient-specific conditions were collected and used to determine the Naranjo score. The most likely associated drugs were the ones that reached the highest Naranjo score concerning the single analyzed AE. Naranjo distinguishes between definitive with a total score ≥9, probable with 5 < total score < 8, possible with 1 < total score < 4 and doubtful with a total score ≤ 0 [21, 22]. Inconclusive evaluations in all steps (i, ii.a, and ii.b) were discussed and finalized by mutual agreement in an expert panel. This panel consisted of four experienced clinical pharmacists. Statistical analysis To ensure comparable patient parameters between the three LTC facilities independent of the allocation to a single facility, main patient parameters were statistically analyzed. For this purpose, a Kruskal Wallis test with pairwise comparison was performed. Analyzed parameters were age, gender, number of diagnoses and number of continuous and on demand drugs, as well as the number of AEs in the patients and the maximum Naranjo score per patient. The data analysis was performed using IBM SPSS Statistics Version 25.0 (IBM Corporation, Armonk, NY, USA) and Microsoft Office Excel 2013 (Microsoft Corporation, Redmond, WA, USA). P-values ≤ 0.05 were considered as statistically significant. Results Patient characteristics In the participating parts of the LTC facilities, 182 patients were potentially available for the study and 154 met the inclusion criteria. From these, 104 patients or their legal guardian gave their informed consent as well as their responsible physician and were enrolled in the study. Patients were mostly female (72.1%) and in median 86 (range: 66–101) years old (Table 1). Patients did not differ between the three LTC facilities according to the following parameters: age (p = 0.311), gender (p = 0.684), number of diagnoses (p = 0.070) and number of continuous (p = 0.629) and on demand drugs (p = 0.911).Table 1 Characteristics of patients included in the study with frequency of documented diagnoses, main ATC classes and main active substances Characteristics Value Patients, total, n 104 Patients in facility of welfare ownership, n (%) 34 (32.7%) Patients in facility of municipal ownership, n (%) 30 (28.8%) Patients in facility of ownership by private association, n (%) 40 (38.5%) Female, n (%) 75 (72.1%) Length of residence (months), median (Q25/Q75; min–max) 31 (12/63; 1–414) Age (years), median (Q25/Q75; min–max) 86 (78/90; 66–101) Documented diagnoses, median (Q25/Q75; min–max) 15 (10/21; 3–35) No. of continuous drugs, median (Q25/Q75; min–max) 8 (6/10; 2–18) No. of on demand medication, median (Q25/Q75; min–max) 2 (1/3; 1–6) Documented diagnosisa Hypertension, n (%) 82 (78.8%) Dementia, n (%) 69 (66.3%) Diabetes, n (%) 41 (39.4%) Heart failure, n (%) 32 (30.8%) Atrial fibrillation, n (%) 32 (30.8%) Renal failure, n (%) 24 (23.1%) Osteoporosis, n (%) 19 (18.3%) Stroke, n (%) 17 (16.3%) Main ATC classesb C (cardiovascular system), n (%) 236 (28.7%) N (nervous system), n (%) 216 (26.3%) A (alimentary tract and metabolism), n (%) 164 (20.0%) B (blood and blood-forming organs), n (%) 68 (8.3%) H (systemic hormonal preparations, excluding sex hormones and insulins), n (%) 27 (3.3%) Main active substancesb Torasemide, n (%) 47 (5.7%) Pantoprazole, n (%) 40 (4.9%) Ramipril, n (%) 35 (4.3%) Acetylsalicylic acid, n (%) 33 (4.0%) Metoprolol, n (%) 23 (2.8%) ATC anatomical therapeutic chemical/defined daily dose classification, Q25/Q75 first and third quartile aOrder is based on the most relevant diagnoses found in literature data to geriatric patients bAccording to the documented continuous drugs (i) AE identification From a total of 104 patients, at least 1 AE was identified in 103 (99.0%). We identified 424 AEs, with a detected median of 4 (Q25/Q75: 2/5, range 1–14) AEs per patient, which equals 2.05 AEs per resident month. The identified AEs and the number of affected patients are shown in Table 2. The system organ classes renal and urinary disorder (87 patients), gastrointestinal disorder (43 patients), skin and subcutaneous tissue disorders (37 patients) were most common in our patient collective. Altogether, 72 different AE categories were detected, 185 AEs were identified in the patient records and 195 AEs by the nurses’ interviews, with 44 AEs in concordance of both methods. We found a significant difference in the detected number of AEs between the observed LTC facilities (p = 0.020). Following the pairwise comparison, we only found differences between the municipal LTC facility with 3 (Q25/Q75: 2/4) AEs and the private LTC facility with a median of 4 (Q25/Q75: 3/6.25) AEs (p = 0.022).Table 2 Identified adverse evnts (n = 424) according to CTCAE and affected patients (n = 104) System organ class Number of identified AEs, n (%) Affected patients, n (%) AE with number of affected patientsa (n) Renal and urinary disorders 88 (20.8) 87 (83.7) Urinary incontinence (87), urinary tract pain (1) Gastrointestinal disorders 55 (13.0) 43 (41.3) Constipation (22), vomiting (16), diarrhea (10), blackened stools (3), nausea (2), lower gastrointestinal bleeding (1), periodontal disease (1) Psychiatric disorders 55 (13.0) 35 (33.7) Confusion (21), restlessness (12), defensive behavior (8), insomnia (5), depression (4), anxiety (2), hallucinations (1), personality change (1), psychiatric disorders—other specify (1) Skin and subcutaneous tissue disorders 50 (11.8) 37 (35.6) Intertrigo (9), dry skin (7), hyperhidrosis (7), skin ulceration (6), local redness (5), pruritus (4), purpura (4), skin and subcutaneous tissue disorders—other specify (3), skin induration (1), urticaria (2), alopecia (1), angioedema (1) Metabolism and nutritional disorders 41 (9.7) 27 (26.0) Hyperglycemia (26), hypoglycemia (15) Musculoskeletal and connective tissue disorders 40 (9.4) 33 (31.7) Arthralgia (14), pain in extremity (12), back pain (6), arthritis (4), musculoskeletal and connective tissue disorders—other specify (3), general muscle weakness (1) Nervous system disorders 39 (9.2) 31 (29.8) Dizziness (11), somnolence (10), headache (4), syncope (3), ataxia (2), cognitive disturbance (2), paresthesia (2), depressed level of consciousness (1), lethargy (1), neuralgia (1), seizure (1), spasticity (1) Injury, poisoning and procedural complications 14 (3.3) 14 (13.5) Fall (14) Vascular disorders 11 (2.6) 11 (10.6) Hematoma (10), flushing (1) Infections and infestations 8 (1.9) 7 (6.7) Skin infection (4), vulval infection (2), conjunctivitis infective (1), stoma site infection (1) General disorders and administration site conditions 7 (1.7) 7 (6.7) Edema limbs (3), pain (3), fatigue (1) Respiratory, thoracic and mediastinal disorders 7 (1.7) 6 (5.8) Dyspnea (4), cough (1), epistaxis (1), respiratory, thoracic and mediastinal disorders—other specify (1) Ear and labyrinth disorders 3 (0.7) 3 (2.9) Hearing impaired (2), tinnitus (1) Cardiac disorders 2 (0.5) 2 (1.9) Chest pain—cardiac (1), palpitations (1) Eye disorders 2 (0.5) 1 (1.0) Blurred vision (1), glaucoma (1) Investigations 2 (0.5) 2 (1.9) Weight gain (1), weight loss (1) AE(s) adverse event(s), CTCAE common terminology criteria for adverse events aMultiple categories per patient possible (ii.a) Review of summary of product characteristics To analyze the concomitantly administered drugs, we assessed 3725 combinations of AEs and corresponding drugs. For this analysis five drug/AE pairs had to be excluded because no information from the SmPC was available (moisturizing eye drops, medical device). Considering every identified AE, patients had a median of 3 potentially causing drugs according to the SmPC, with a range from 0 to 11 drugs (Q25/Q75: 2/4; details in Fig. 1). The most frequently (n ≥ 10) detected AEs and the affected system organ classes are shown in Table 3. The ATC classes prescribed most often (C, N, A, B, H) were frequently among the potentially causing drugs for the most common system organ classes (Fig. 2).Fig. 1 Number of detected adverse events versus number of potentially causing drugs according Summary of Product Charactetistics. AE(s) adverse event(s), SmPC summary of products characteristics Table 3 Median number of potentially causing drugs according Summary of Product Charactersitics and corresponding Naranjo Score per patient (n = 104) for the most frequently detected (≥10) adverse events (AEs) and for their corresponding System organ classes (all 424 detected AE included) System organ class and ... most frequent AE Number (n) Median number of potentially causing drugs per patient [range] Median Naranjo score [range] Renal and urinary disorders 88 2 [0–5] 0 [−1–2] ... Urinary incontinence 87 2 [0–5] 0 [−1–2] Gastrointestinal disorders 55 5 [0–11] 2 [0–4] ... Constipation 22 5 [0–10] 0 [0–3] ... Vomiting 16 7.5 [2–10] 2 [0–4] ... Diarrhea 10 4.5 [2–11] 3 [0–3] Psychiatric disorders 55 3 [0–7] 0 [−1–9] ... Confusion 21 3 [1–7] 1 [0–8] ... Restlessness 12 3 [0–4] 0 [−1–3] Metabolism and nutrition disorders 41 3 [0–5] 1 [0–5] ... Hyperglycemia 26 3 [0–4] 1 [0–1] ... Hypoglycemia 15 3 [1–5] 4 [2–5] Musculoskeletal and connective tissue disorders 40 3 [0–8] 2 [−1–3] ... Arthralgia 14 3 [0–6] 1 [−1–3] ... Pain in extremity 12 2 [1–8] 2 [0–3] Nervous system disorders 39 4 [0–10] 1 [−1–7] ... Dizziness 11 5 [1–10] 2 [0–7] ... Somnolence 10 4 [3–7] 3 [0–5] Injury, poisoning and procedural complications 14 6 [3–11] 2 [0–3] ... Fall 14 6 [3–11] 2 [0–3] Vascular disorders 11 1 [0–3] 1 [0–3] ... Hematoma 10 1 [0–3] 0.5 [0–2] AE(s) Adverse Event(s), SmPC Summary of product characteristics Fig. 2 Potentially causing drugs according Summary of Product Characteristics differentiated into the ATC classes for the most frequently detected system organ classes. AE(s) adverse event(s), ATC anatomical therapeutic chemical/defined daily dose classification, CTCAE common criteria for the terminology of adverse events, SmPC summary of products characteristics. ATC classes: A: alimentary tract and metabolism, B: blood and blood forming organs, C: cardiovascular system, H: systemic hormonal preparations, excluding sex hormones and insulins, N: nervous system (ii.b) Causality assessment according to the Naranjo algorithm All 3730 drug/AE pairs were included in the causality assessment. From the 424 identified AEs, 198 (46.9%) were classified as ADR with “doubtful”, 218 (51.2%) “possible”, 7 (1.7%) “probable”, and 1 (0.2%) “definitive” cause (Table 4). We found no significant differences in the maximum Naranjo scores per patient between the three LTC facilities (p = 0.964). On the basis of 424 detected AEs, only 1 drug in 84 AEs (19.8%) and several drugs in 340 AEs (80.2%) reached the highest score (Table 4). According to Naranjo these need to be considered as the most likely causing drug(s).Table 4 Results of the adverse event drug causality assessment according to the Naranjo algorithm Naranjo Score per AE Identified AE [n] Number of affected patientsa, [n] Classification Identified AE per class, n (%) Number of AE with one/several highest Naranjo drug(s) [n] −1 22 17 Doubtful 199 (46.9) 38/161 0 177 92 1 68 48 Possible 217 (51.2) 38/179 2 90 51 3 41 30 4 18 18 5 3 3 Probable 7 (1.7) 7/0 6 2 2 7 1 1 8 1 1 9 1 1 Definitive 1 (0.2) 1/0 AE(s) adverse event(s) aSeveral AEs per patient possible The probable and definitive ADRs were as follows: angioedema (severity grade according to CTCAE 4) induced by enalapril, urticaria (grade 2) induced by amoxicillin and clavulanic acid, hypoglycemia (grade 1) induced by insulin glargine, paresthesia (grade 2) induced by tapentadol and a complex case of occurring hallucinations (grade 3) in combination with confusion (grade 3), dizziness (grade 3) and somnolence (grade 3, in total 4 detected AEs) which were attributable to digitoxin (highest Naranjo score). In this case, the patient also received high doses of oxycodone and duloxetine. It can be seen as a mixed intoxication based on the hospital report. In the algorithm, digitoxin reached a one-point higher score than oxycodone/duloxetine because measurement of the increased blood level was available only for digitoxin. All of the detected AEs and ADRs were managed adequately by the nurses, for example, by informing a physician or arranging a hospital admission for the affected patient. Thus, no further action was required due to this study. Discussion In our study we addressed AEs in geriatric patients living in LTC facilities. We assessed which type of AEs occurred and also investigated potential additive effects of polypharmacy. With nearly all (99%) patients affected by AEs, we demonstrated the relevance of this topic. We found the identified AEs potentially caused by up to 11 different administered drugs. The Naranjo algorithm showed at least possible drug causes in half of these AEs. Thereby, multiple drugs were equally likely involved 80% of the time. Our results point out that AEs should be systematically recorded in routine practice in LTC facilities. In order to prevent ADRs, additive effects need to be considered in any strategies developed. Prevalence of AE and ADR in LTC residents More than half of our identified AEs could be associated with drug use. Our rate of probable and definitive ADRs was similar to other studies in the LTC setting (0.04 vs. up to 0.10 ADRs per observed resident month), although studies should be compared with caution [9, 23]. Nevertheless, the causality assessment leaves us with a high number of possible ADRs. Especially for AEs which were ongoing for a longer period, causality assessment was challenging in the routine setting. Information to evaluate the exact temporal connection between AE and drug use was frequently missing and therefore could have led to lower Naranjo scores. To resolve this problem, a regular and structured routine assessment of AEs and potentially causing drugs might increase the chance to identify ADRs and protect patients from the consequences. Our overall rate of identified AEs was higher than results seen in other studies (2.05 AEs vs 0.03–0.12 per observed resident month) [9, 23]. This indicates that we identified a noticeable amount of the general symptom burden of LTC residents that results from underlying diseases or age-related changes. This is consistent with the fact that incontinence, pain, sleep disorders and psychopathological symptoms are widely found in LTC residents [24]. Therefore, a regular routine AE assessment can support ADR detection as well as structured symptom evaluation. Additive effects of polypharmacy The suspected AE was listed as an ADR in the respective SmPC in a median of 3 and up to 11 administered drugs per patient. In 80% of all identified AEs, various drugs reached the highest Naranjo score simultaneously. This means that they were equally likely to cause the AE. This coincidence can increase the chance of AE occurrence independently from single causality scores. This result also raises the question whether ADRs resulting from additive effects have been underestimated. In cases with “probable” or “definitive” ADRs (Naranjo ≥5), we found results from only one drug with the highest Naranjo score; however, in four of these AEs, the drug with the highest Naranjo (digitoxin) was only part of a mixed intoxication with duloxetine and oxycodone based on the hospital report for the affected patient. In this case, the sole consideration of the causality assessment could mask an additive effect of at least 3 concomitantly given drugs. This shows that additive effects need to be considered in every detected AE independently from the single causality. Besides ADRs from well-known drug classes (e.g. vascular ADRs from drugs affecting blood and blood-forming organs), in a substantial amount of AEs, we found involvement of varying ATC-classes that are less familiar (e.g. nervous system ADRs in drugs affecting the cardiovascular system). This underlines the complexity of geriatric patient treatment and the need for interdisciplinary medication reviews that include an assessment of drug-related problems, such as drug-drug interactions, potentially inappropriate medication, as well as ADRs [25, 26]. In routine care, however, potential additive effects are often not taken into account. In particular, new and unclear symptoms could be misinterpreted as new diseases and sometimes even lead to prescribing cascades [27, 28]. Implications for practice Firstly, our study shows the need for a good data base and a regular routine assessment of AEs that occur in LTC facilities. We found a very low concordance rate of only 10% between AEs detected in nurses’ interviews and those mentioned in the patient record analysis. This demonstrates the potential of information loss in LTC facilities due to heterogeneous and incomplete AE documentation [29]. It also indicates the potential of recall bias in the nurses. The identification of every occurring AE allows a better assessment of simultaneously occurring events. We found in our study, for example, a combination of vomiting and diarrhea that indicated an infection rather than an ADR. Furthermore, the information on patients’ current symptoms contributes to appropriate proposals for medication changes in cases of identified ADRs. Secondly, our results support the development of strategies with improved consideration of the additive effects of polypharmacy. Combining an AE assessment with structured medication reviews improves the drug cause analysis of AEs as well as the detection and interpretation of drug-related problems. Ongoing prospective evaluation of AEs and potential drug-related causes contributes to prevent patients from experiencing negative events. This process could be further accelerated by electronic assistance. Electronic documentation of AEs and computer-assisted signal detection of ADRs can support problem solving in a narrow timeframe since physicians and pharmacists are usually not permanently present in the LTC facilities [11, 30]. Database-supported comparison of the events with patients’ medication can assist pharmacists in a comprehensive medication review. Currently, such electronic solutions are rarely used in the LTC setting in Germany. They could also support future research by providing information on the additive effects of various combined drugs and underlying diseases. Thirdly, our data suggest the need for improvement in interdisciplinary communication in LTC facilities. In interprofessional teams with nurses, pharmacists and physicians, systematic information about AEs, medication reviews and actual health conditions could be transmitted more effectively in patient-orientated practice. Conclusion Nearly every long-term care resident suffered from adverse events (AEs), with half of them at least possibly caused by drugs. In four fifths of these AEs, several concomitantly given drugs were equally associated causes. Therefore, potential additive effects need to be considered independently from single causality and should be more focused in further research. A routinely implemented structured search for AEs and additive effects of polypharmacy contributes to medication reviews and interdisciplinary collaboration and will help to meet the needs of this complex patient collective and to protect them from negative consequences. Acknowledgements We thank all participating co-workers of the LTC facilities and the attending physicians for their support. Furthermore, we thank PhD Johanna Freyer for her assistance with the ethics approval and Katharine Worthington for language editing. Funding A co-worker of the study (Monika Lexow) was financially supported in part by the Lesmueller Foundation, Munich, Germany, the German Pharmacist Foundation, Berlin, Germany and the Pharmacist Foundation Westfalen-Lippe, Münster, Germany. Author Contribution Conceptualization: Monika Lexow, Kathrin Wernecke, Ralf Sulzer, Thilo Bertsche, Susanne Schiek; methodology: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; formal analysis and investigation: Kathrin Wernecke, Susanne Schiek; investigation: Monika Lexow, Kathrin Wernecke; writing, original draft preparation: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; writing, review and editing: Monika Lexow, Kathrin Wernecke, Ralf Sultzer, Gordian L. Schmid, Thilo Bertsche, Susanne Schiek; visualization: Monika Lexow, Kathrin Wernecke, Susanne Schiek; supervision: Thilo Bertsche, Susanne Schiek; project administration: Monika Lexow; funding acquisition: Thilo Bertsche, Susanne Schiek. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with ethical guidelines Conflict of interest M. Lexow, K. Wernecke, G.L. Schmid, R. Sultzer, T. Bertsche, and S. Schiek declare that they have no competing interests. Ethical standards All procedures performed in this study involving human participants were approved both by the ethics committee of the Faculty of Medicine of Leipzig University as well as the ethics committee of the State Chamber of Physicians of Saxony (reference: 231/13-ff and EK-allg-26/14-1) and have been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All participants gave their informed consent prior to inclusion in the study. The authors M. Lexow and K. Wernecke contributed equally to the manuscript. The authors T. Bertsche and S. Schiek contributed equally to the manuscript. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Recovering
ReactionOutcome
CC BY
33090261
20,027,693
2021-08
What was the outcome of reaction 'Somnolence'?
Considering additive effects of polypharmacy : Analysis of adverse events in geriatric patients in long-term care facilities. BACKGROUND Potential additive effects of polypharmacy are rarely considered in adverse events of geriatric patients living in long-term care facilities. Our aim, therefore, was to identify adverse events in this setting and to assess plausible concomitant drug causes. METHODS A cross-sectional observational study was performed in three facilities as follows: (i) adverse event identification: we structurally identified adverse events using nurses' interviews and chart review. (ii) Analysis of the concomitantly administered drugs per patient was performed in two ways: (ii.a) a review of summary of product characteristics for listed adverse drug reactions to identify possible causing drugs and (ii.b) a causality assessment according to Naranjo algorithm. RESULTS (i) We found 424 adverse events with a median of 4 per patient (range 1-14) in 103 of the 104 enrolled patients (99%). (ii.a) We identified a median of 3 drugs (range 0-11) with actually occurring adverse events listed as an adverse drug reaction in the summary of product characteristics. (ii.b) Causality was classified in 198 (46.9%) of adverse events as "doubtful," in 218 (51.2%) as "possible," in 7 (1.7%) as "probable," and in 1 (0.2%) adverse event as a "definitive" cause of the administered drugs. In 340 (80.2%) of all identified adverse events several drugs simultaneously reached the highest respective Naranjo score. CONCLUSIONS Patients in long-term facilities frequently suffer from many adverse events. Concomitantly administered drugs have to be frequently considered as plausible causes for adverse events. These additive effects of drugs should be more focused in patient care and research. Introduction Geriatric patients in long-term care (LTC) facilities are multimorbid and, therefore, suffer from many (non)specific symptoms and geriatric syndromes [1]. Disease-related symptoms should be distinguished from adverse drug reactions (ADR) that result from drug therapy [2]. The latter can lead to hospital admissions and have a considerable impact on morbidity and mortality with high costs in the health care system [3–5]. Polypharmacy makes a significant contribution to the clinical consequences deriving from ADRs in geriatric patients [6]. For this reason, a structured analysis of adverse events (AE) and drug-related causes in these patients is of high interest for routine care. Distinguishing whether an observed AE was caused by a disease (i.e. symptom) or by a drug (ADR) poses a challenge for healthcare professionals [7, 8]. The correct attribution is required for appropriate treatment strategies but can result only from structured detection, analysis and classification. Geriatric patients are frequently cognitively impaired or suffer from speech or hearing disorders. Hence, information provided by the patients is often insufficient. In LTC facilities, therefore, chart documentation and nurses’ interviews are the most valuable sources for AE detection [9, 10]. So far, no specific method exists to analyze and classify AE in LTC facility residents with polypharmacy. The Naranjo algorithm has previously been used for causality assessment in this collective [11, 12]. It allows a detailed assessment of every detected AE and every single administered drug. This algorithm provides further information on drug-related causes when combined with established methods for patient safety, such as drug-drug interactions and potentially inappropriate medications [13]. Causality scores like the Naranjo score, however, do not consider simultaneously contributing drugs. For some ADRs, it has been shown that the number of specific drugs causes their clinical manifestations. For example, patients are exposed to an increased risk of falling when they take two or more drugs which increase the risk of falling [14]. Concerning anticholinergic ADRs, it is common to calculate an anticholinergic burden to quantify the risk for an adverse outcome [15]. Little is known, however, about additive drug effects in other events. Therefore, data about potential additive effects in this vulnerable patient collective are of great interest for routine care. The aim of this study was to identify AEs occurring in LTC facility patients and to assess plausible concomitant drug causes. Patients, material and methods Definitions We defined an AE as an outcome that occurs while a patient is taking a drug, but is not or not necessarily attributable to it and an ADR as an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product [16]. We used the term drug not only for the effective substance but for the whole product prescribed in the medication chart of the patient. A drug therefore could contain more than one active substance. We considered all drugs administered to the patient during the acquisition period. Continuous and on-demand medications were assessed separately because the temporal relationship between AE and administration of the drug could be different in that case. Participants and setting We conducted a cross-sectional observation study in three LTC facilities in Germany. After written informed consent of the residents or their legal representative and the responsible general practitioner, residents in the participating LTC facilities were enrolled in the study. We included residents of facilities with different ownerships (welfare, municipal or private associations) to approach a representative sample of 100 residents. Inclusion criteria were: informed consent, age ≥65 years, long-term/chronic medicines ≥3 and multimorbidity with ≥3 comorbidities at the time of recruitment, more than 8 weeks stay in the LTC facility, and a life expectancy of more than 6 months according to nurses’ present information. The study was conducted over a time period of 10 months. Study design and data collection We conducted a structured analysis of AEs. (i) AE identification We used two complement sources of information for our structured data collection: Firstly, an interview about individual AEs with nurses involved in daily care and, secondly, a review of residents’ records (electronic and chart documentation, laboratory values) for documented events and their temporal occurrence. To ensure standardized identification of AEs, a checklist of events was applied to both methods. The listed events comprised the most relevant AEs or ADRs for geriatric patients and LTC residents based on the literature [17–19]: blackened stool, bleeding/hematoma, confusion/disorientation, constipation, depression/anxiety, diarrhea, dizziness/vertigo, dry mouth, ear disorders, eye disorders, falls, hallucination, hyperglycemia/hypoglycemia, hyperhidrosis, hyperkalemia/hypokalemia, hypernatremia/hyponatremia, nausea, pain, restlessness, skin disorders/pruritus, insomnia, urinary incontinence, vomiting (in alphabetical order). Additional relevant reported or documented events were collected as well. We considered reported and documented symptoms during a time period of the prior 30 days (considered as 1 resident month) for new and continuous symptoms. Data collection was performed at two measurement points per patient at intervals of 6–8 weeks by two clinical pharmacists. All detected AEs and the corresponding system organ class were classified based on the common terminology criteria for adverse events (CTCAE) [20]. (iia) Review of summary of product characteristics We systematically collected data from the medical charts (continuous drugs, on demand drugs and their frequency of use, date of first administration). We checked all summaries of product characteristics (SmPCs) of the actually administered drugs for listed ADRs. A drug would be considered as “potentially causing” if the listed ADR in the SmPC represented a synonym for the detected AE or possibly caused it (e.g. dizziness in cases of falls). For the analysis of additive effects, we counted the number of potentially causing drugs. Prescribed drugs were characterized by their code in the anatomical therapeutic chemical classification system (ATC code). (iib) Causality assessment according to the Naranjo algorithm We used the Naranjo algorithm for causality assessment. All further relevant information, such as the duration of the AE, underlying diseases, clinical consequences (e.g. from hospital report), laboratory values, and patient-specific conditions were collected and used to determine the Naranjo score. The most likely associated drugs were the ones that reached the highest Naranjo score concerning the single analyzed AE. Naranjo distinguishes between definitive with a total score ≥9, probable with 5 < total score < 8, possible with 1 < total score < 4 and doubtful with a total score ≤ 0 [21, 22]. Inconclusive evaluations in all steps (i, ii.a, and ii.b) were discussed and finalized by mutual agreement in an expert panel. This panel consisted of four experienced clinical pharmacists. Statistical analysis To ensure comparable patient parameters between the three LTC facilities independent of the allocation to a single facility, main patient parameters were statistically analyzed. For this purpose, a Kruskal Wallis test with pairwise comparison was performed. Analyzed parameters were age, gender, number of diagnoses and number of continuous and on demand drugs, as well as the number of AEs in the patients and the maximum Naranjo score per patient. The data analysis was performed using IBM SPSS Statistics Version 25.0 (IBM Corporation, Armonk, NY, USA) and Microsoft Office Excel 2013 (Microsoft Corporation, Redmond, WA, USA). P-values ≤ 0.05 were considered as statistically significant. Results Patient characteristics In the participating parts of the LTC facilities, 182 patients were potentially available for the study and 154 met the inclusion criteria. From these, 104 patients or their legal guardian gave their informed consent as well as their responsible physician and were enrolled in the study. Patients were mostly female (72.1%) and in median 86 (range: 66–101) years old (Table 1). Patients did not differ between the three LTC facilities according to the following parameters: age (p = 0.311), gender (p = 0.684), number of diagnoses (p = 0.070) and number of continuous (p = 0.629) and on demand drugs (p = 0.911).Table 1 Characteristics of patients included in the study with frequency of documented diagnoses, main ATC classes and main active substances Characteristics Value Patients, total, n 104 Patients in facility of welfare ownership, n (%) 34 (32.7%) Patients in facility of municipal ownership, n (%) 30 (28.8%) Patients in facility of ownership by private association, n (%) 40 (38.5%) Female, n (%) 75 (72.1%) Length of residence (months), median (Q25/Q75; min–max) 31 (12/63; 1–414) Age (years), median (Q25/Q75; min–max) 86 (78/90; 66–101) Documented diagnoses, median (Q25/Q75; min–max) 15 (10/21; 3–35) No. of continuous drugs, median (Q25/Q75; min–max) 8 (6/10; 2–18) No. of on demand medication, median (Q25/Q75; min–max) 2 (1/3; 1–6) Documented diagnosisa Hypertension, n (%) 82 (78.8%) Dementia, n (%) 69 (66.3%) Diabetes, n (%) 41 (39.4%) Heart failure, n (%) 32 (30.8%) Atrial fibrillation, n (%) 32 (30.8%) Renal failure, n (%) 24 (23.1%) Osteoporosis, n (%) 19 (18.3%) Stroke, n (%) 17 (16.3%) Main ATC classesb C (cardiovascular system), n (%) 236 (28.7%) N (nervous system), n (%) 216 (26.3%) A (alimentary tract and metabolism), n (%) 164 (20.0%) B (blood and blood-forming organs), n (%) 68 (8.3%) H (systemic hormonal preparations, excluding sex hormones and insulins), n (%) 27 (3.3%) Main active substancesb Torasemide, n (%) 47 (5.7%) Pantoprazole, n (%) 40 (4.9%) Ramipril, n (%) 35 (4.3%) Acetylsalicylic acid, n (%) 33 (4.0%) Metoprolol, n (%) 23 (2.8%) ATC anatomical therapeutic chemical/defined daily dose classification, Q25/Q75 first and third quartile aOrder is based on the most relevant diagnoses found in literature data to geriatric patients bAccording to the documented continuous drugs (i) AE identification From a total of 104 patients, at least 1 AE was identified in 103 (99.0%). We identified 424 AEs, with a detected median of 4 (Q25/Q75: 2/5, range 1–14) AEs per patient, which equals 2.05 AEs per resident month. The identified AEs and the number of affected patients are shown in Table 2. The system organ classes renal and urinary disorder (87 patients), gastrointestinal disorder (43 patients), skin and subcutaneous tissue disorders (37 patients) were most common in our patient collective. Altogether, 72 different AE categories were detected, 185 AEs were identified in the patient records and 195 AEs by the nurses’ interviews, with 44 AEs in concordance of both methods. We found a significant difference in the detected number of AEs between the observed LTC facilities (p = 0.020). Following the pairwise comparison, we only found differences between the municipal LTC facility with 3 (Q25/Q75: 2/4) AEs and the private LTC facility with a median of 4 (Q25/Q75: 3/6.25) AEs (p = 0.022).Table 2 Identified adverse evnts (n = 424) according to CTCAE and affected patients (n = 104) System organ class Number of identified AEs, n (%) Affected patients, n (%) AE with number of affected patientsa (n) Renal and urinary disorders 88 (20.8) 87 (83.7) Urinary incontinence (87), urinary tract pain (1) Gastrointestinal disorders 55 (13.0) 43 (41.3) Constipation (22), vomiting (16), diarrhea (10), blackened stools (3), nausea (2), lower gastrointestinal bleeding (1), periodontal disease (1) Psychiatric disorders 55 (13.0) 35 (33.7) Confusion (21), restlessness (12), defensive behavior (8), insomnia (5), depression (4), anxiety (2), hallucinations (1), personality change (1), psychiatric disorders—other specify (1) Skin and subcutaneous tissue disorders 50 (11.8) 37 (35.6) Intertrigo (9), dry skin (7), hyperhidrosis (7), skin ulceration (6), local redness (5), pruritus (4), purpura (4), skin and subcutaneous tissue disorders—other specify (3), skin induration (1), urticaria (2), alopecia (1), angioedema (1) Metabolism and nutritional disorders 41 (9.7) 27 (26.0) Hyperglycemia (26), hypoglycemia (15) Musculoskeletal and connective tissue disorders 40 (9.4) 33 (31.7) Arthralgia (14), pain in extremity (12), back pain (6), arthritis (4), musculoskeletal and connective tissue disorders—other specify (3), general muscle weakness (1) Nervous system disorders 39 (9.2) 31 (29.8) Dizziness (11), somnolence (10), headache (4), syncope (3), ataxia (2), cognitive disturbance (2), paresthesia (2), depressed level of consciousness (1), lethargy (1), neuralgia (1), seizure (1), spasticity (1) Injury, poisoning and procedural complications 14 (3.3) 14 (13.5) Fall (14) Vascular disorders 11 (2.6) 11 (10.6) Hematoma (10), flushing (1) Infections and infestations 8 (1.9) 7 (6.7) Skin infection (4), vulval infection (2), conjunctivitis infective (1), stoma site infection (1) General disorders and administration site conditions 7 (1.7) 7 (6.7) Edema limbs (3), pain (3), fatigue (1) Respiratory, thoracic and mediastinal disorders 7 (1.7) 6 (5.8) Dyspnea (4), cough (1), epistaxis (1), respiratory, thoracic and mediastinal disorders—other specify (1) Ear and labyrinth disorders 3 (0.7) 3 (2.9) Hearing impaired (2), tinnitus (1) Cardiac disorders 2 (0.5) 2 (1.9) Chest pain—cardiac (1), palpitations (1) Eye disorders 2 (0.5) 1 (1.0) Blurred vision (1), glaucoma (1) Investigations 2 (0.5) 2 (1.9) Weight gain (1), weight loss (1) AE(s) adverse event(s), CTCAE common terminology criteria for adverse events aMultiple categories per patient possible (ii.a) Review of summary of product characteristics To analyze the concomitantly administered drugs, we assessed 3725 combinations of AEs and corresponding drugs. For this analysis five drug/AE pairs had to be excluded because no information from the SmPC was available (moisturizing eye drops, medical device). Considering every identified AE, patients had a median of 3 potentially causing drugs according to the SmPC, with a range from 0 to 11 drugs (Q25/Q75: 2/4; details in Fig. 1). The most frequently (n ≥ 10) detected AEs and the affected system organ classes are shown in Table 3. The ATC classes prescribed most often (C, N, A, B, H) were frequently among the potentially causing drugs for the most common system organ classes (Fig. 2).Fig. 1 Number of detected adverse events versus number of potentially causing drugs according Summary of Product Charactetistics. AE(s) adverse event(s), SmPC summary of products characteristics Table 3 Median number of potentially causing drugs according Summary of Product Charactersitics and corresponding Naranjo Score per patient (n = 104) for the most frequently detected (≥10) adverse events (AEs) and for their corresponding System organ classes (all 424 detected AE included) System organ class and ... most frequent AE Number (n) Median number of potentially causing drugs per patient [range] Median Naranjo score [range] Renal and urinary disorders 88 2 [0–5] 0 [−1–2] ... Urinary incontinence 87 2 [0–5] 0 [−1–2] Gastrointestinal disorders 55 5 [0–11] 2 [0–4] ... Constipation 22 5 [0–10] 0 [0–3] ... Vomiting 16 7.5 [2–10] 2 [0–4] ... Diarrhea 10 4.5 [2–11] 3 [0–3] Psychiatric disorders 55 3 [0–7] 0 [−1–9] ... Confusion 21 3 [1–7] 1 [0–8] ... Restlessness 12 3 [0–4] 0 [−1–3] Metabolism and nutrition disorders 41 3 [0–5] 1 [0–5] ... Hyperglycemia 26 3 [0–4] 1 [0–1] ... Hypoglycemia 15 3 [1–5] 4 [2–5] Musculoskeletal and connective tissue disorders 40 3 [0–8] 2 [−1–3] ... Arthralgia 14 3 [0–6] 1 [−1–3] ... Pain in extremity 12 2 [1–8] 2 [0–3] Nervous system disorders 39 4 [0–10] 1 [−1–7] ... Dizziness 11 5 [1–10] 2 [0–7] ... Somnolence 10 4 [3–7] 3 [0–5] Injury, poisoning and procedural complications 14 6 [3–11] 2 [0–3] ... Fall 14 6 [3–11] 2 [0–3] Vascular disorders 11 1 [0–3] 1 [0–3] ... Hematoma 10 1 [0–3] 0.5 [0–2] AE(s) Adverse Event(s), SmPC Summary of product characteristics Fig. 2 Potentially causing drugs according Summary of Product Characteristics differentiated into the ATC classes for the most frequently detected system organ classes. AE(s) adverse event(s), ATC anatomical therapeutic chemical/defined daily dose classification, CTCAE common criteria for the terminology of adverse events, SmPC summary of products characteristics. ATC classes: A: alimentary tract and metabolism, B: blood and blood forming organs, C: cardiovascular system, H: systemic hormonal preparations, excluding sex hormones and insulins, N: nervous system (ii.b) Causality assessment according to the Naranjo algorithm All 3730 drug/AE pairs were included in the causality assessment. From the 424 identified AEs, 198 (46.9%) were classified as ADR with “doubtful”, 218 (51.2%) “possible”, 7 (1.7%) “probable”, and 1 (0.2%) “definitive” cause (Table 4). We found no significant differences in the maximum Naranjo scores per patient between the three LTC facilities (p = 0.964). On the basis of 424 detected AEs, only 1 drug in 84 AEs (19.8%) and several drugs in 340 AEs (80.2%) reached the highest score (Table 4). According to Naranjo these need to be considered as the most likely causing drug(s).Table 4 Results of the adverse event drug causality assessment according to the Naranjo algorithm Naranjo Score per AE Identified AE [n] Number of affected patientsa, [n] Classification Identified AE per class, n (%) Number of AE with one/several highest Naranjo drug(s) [n] −1 22 17 Doubtful 199 (46.9) 38/161 0 177 92 1 68 48 Possible 217 (51.2) 38/179 2 90 51 3 41 30 4 18 18 5 3 3 Probable 7 (1.7) 7/0 6 2 2 7 1 1 8 1 1 9 1 1 Definitive 1 (0.2) 1/0 AE(s) adverse event(s) aSeveral AEs per patient possible The probable and definitive ADRs were as follows: angioedema (severity grade according to CTCAE 4) induced by enalapril, urticaria (grade 2) induced by amoxicillin and clavulanic acid, hypoglycemia (grade 1) induced by insulin glargine, paresthesia (grade 2) induced by tapentadol and a complex case of occurring hallucinations (grade 3) in combination with confusion (grade 3), dizziness (grade 3) and somnolence (grade 3, in total 4 detected AEs) which were attributable to digitoxin (highest Naranjo score). In this case, the patient also received high doses of oxycodone and duloxetine. It can be seen as a mixed intoxication based on the hospital report. In the algorithm, digitoxin reached a one-point higher score than oxycodone/duloxetine because measurement of the increased blood level was available only for digitoxin. All of the detected AEs and ADRs were managed adequately by the nurses, for example, by informing a physician or arranging a hospital admission for the affected patient. Thus, no further action was required due to this study. Discussion In our study we addressed AEs in geriatric patients living in LTC facilities. We assessed which type of AEs occurred and also investigated potential additive effects of polypharmacy. With nearly all (99%) patients affected by AEs, we demonstrated the relevance of this topic. We found the identified AEs potentially caused by up to 11 different administered drugs. The Naranjo algorithm showed at least possible drug causes in half of these AEs. Thereby, multiple drugs were equally likely involved 80% of the time. Our results point out that AEs should be systematically recorded in routine practice in LTC facilities. In order to prevent ADRs, additive effects need to be considered in any strategies developed. Prevalence of AE and ADR in LTC residents More than half of our identified AEs could be associated with drug use. Our rate of probable and definitive ADRs was similar to other studies in the LTC setting (0.04 vs. up to 0.10 ADRs per observed resident month), although studies should be compared with caution [9, 23]. Nevertheless, the causality assessment leaves us with a high number of possible ADRs. Especially for AEs which were ongoing for a longer period, causality assessment was challenging in the routine setting. Information to evaluate the exact temporal connection between AE and drug use was frequently missing and therefore could have led to lower Naranjo scores. To resolve this problem, a regular and structured routine assessment of AEs and potentially causing drugs might increase the chance to identify ADRs and protect patients from the consequences. Our overall rate of identified AEs was higher than results seen in other studies (2.05 AEs vs 0.03–0.12 per observed resident month) [9, 23]. This indicates that we identified a noticeable amount of the general symptom burden of LTC residents that results from underlying diseases or age-related changes. This is consistent with the fact that incontinence, pain, sleep disorders and psychopathological symptoms are widely found in LTC residents [24]. Therefore, a regular routine AE assessment can support ADR detection as well as structured symptom evaluation. Additive effects of polypharmacy The suspected AE was listed as an ADR in the respective SmPC in a median of 3 and up to 11 administered drugs per patient. In 80% of all identified AEs, various drugs reached the highest Naranjo score simultaneously. This means that they were equally likely to cause the AE. This coincidence can increase the chance of AE occurrence independently from single causality scores. This result also raises the question whether ADRs resulting from additive effects have been underestimated. In cases with “probable” or “definitive” ADRs (Naranjo ≥5), we found results from only one drug with the highest Naranjo score; however, in four of these AEs, the drug with the highest Naranjo (digitoxin) was only part of a mixed intoxication with duloxetine and oxycodone based on the hospital report for the affected patient. In this case, the sole consideration of the causality assessment could mask an additive effect of at least 3 concomitantly given drugs. This shows that additive effects need to be considered in every detected AE independently from the single causality. Besides ADRs from well-known drug classes (e.g. vascular ADRs from drugs affecting blood and blood-forming organs), in a substantial amount of AEs, we found involvement of varying ATC-classes that are less familiar (e.g. nervous system ADRs in drugs affecting the cardiovascular system). This underlines the complexity of geriatric patient treatment and the need for interdisciplinary medication reviews that include an assessment of drug-related problems, such as drug-drug interactions, potentially inappropriate medication, as well as ADRs [25, 26]. In routine care, however, potential additive effects are often not taken into account. In particular, new and unclear symptoms could be misinterpreted as new diseases and sometimes even lead to prescribing cascades [27, 28]. Implications for practice Firstly, our study shows the need for a good data base and a regular routine assessment of AEs that occur in LTC facilities. We found a very low concordance rate of only 10% between AEs detected in nurses’ interviews and those mentioned in the patient record analysis. This demonstrates the potential of information loss in LTC facilities due to heterogeneous and incomplete AE documentation [29]. It also indicates the potential of recall bias in the nurses. The identification of every occurring AE allows a better assessment of simultaneously occurring events. We found in our study, for example, a combination of vomiting and diarrhea that indicated an infection rather than an ADR. Furthermore, the information on patients’ current symptoms contributes to appropriate proposals for medication changes in cases of identified ADRs. Secondly, our results support the development of strategies with improved consideration of the additive effects of polypharmacy. Combining an AE assessment with structured medication reviews improves the drug cause analysis of AEs as well as the detection and interpretation of drug-related problems. Ongoing prospective evaluation of AEs and potential drug-related causes contributes to prevent patients from experiencing negative events. This process could be further accelerated by electronic assistance. Electronic documentation of AEs and computer-assisted signal detection of ADRs can support problem solving in a narrow timeframe since physicians and pharmacists are usually not permanently present in the LTC facilities [11, 30]. Database-supported comparison of the events with patients’ medication can assist pharmacists in a comprehensive medication review. Currently, such electronic solutions are rarely used in the LTC setting in Germany. They could also support future research by providing information on the additive effects of various combined drugs and underlying diseases. Thirdly, our data suggest the need for improvement in interdisciplinary communication in LTC facilities. In interprofessional teams with nurses, pharmacists and physicians, systematic information about AEs, medication reviews and actual health conditions could be transmitted more effectively in patient-orientated practice. Conclusion Nearly every long-term care resident suffered from adverse events (AEs), with half of them at least possibly caused by drugs. In four fifths of these AEs, several concomitantly given drugs were equally associated causes. Therefore, potential additive effects need to be considered independently from single causality and should be more focused in further research. A routinely implemented structured search for AEs and additive effects of polypharmacy contributes to medication reviews and interdisciplinary collaboration and will help to meet the needs of this complex patient collective and to protect them from negative consequences. Acknowledgements We thank all participating co-workers of the LTC facilities and the attending physicians for their support. Furthermore, we thank PhD Johanna Freyer for her assistance with the ethics approval and Katharine Worthington for language editing. Funding A co-worker of the study (Monika Lexow) was financially supported in part by the Lesmueller Foundation, Munich, Germany, the German Pharmacist Foundation, Berlin, Germany and the Pharmacist Foundation Westfalen-Lippe, Münster, Germany. Author Contribution Conceptualization: Monika Lexow, Kathrin Wernecke, Ralf Sulzer, Thilo Bertsche, Susanne Schiek; methodology: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; formal analysis and investigation: Kathrin Wernecke, Susanne Schiek; investigation: Monika Lexow, Kathrin Wernecke; writing, original draft preparation: Monika Lexow, Kathrin Wernecke, Thilo Bertsche, Susanne Schiek; writing, review and editing: Monika Lexow, Kathrin Wernecke, Ralf Sultzer, Gordian L. Schmid, Thilo Bertsche, Susanne Schiek; visualization: Monika Lexow, Kathrin Wernecke, Susanne Schiek; supervision: Thilo Bertsche, Susanne Schiek; project administration: Monika Lexow; funding acquisition: Thilo Bertsche, Susanne Schiek. Funding Open Access funding enabled and organized by Projekt DEAL. Compliance with ethical guidelines Conflict of interest M. Lexow, K. Wernecke, G.L. Schmid, R. Sultzer, T. Bertsche, and S. Schiek declare that they have no competing interests. Ethical standards All procedures performed in this study involving human participants were approved both by the ethics committee of the Faculty of Medicine of Leipzig University as well as the ethics committee of the State Chamber of Physicians of Saxony (reference: 231/13-ff and EK-allg-26/14-1) and have been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All participants gave their informed consent prior to inclusion in the study. The authors M. Lexow and K. Wernecke contributed equally to the manuscript. The authors T. Bertsche and S. Schiek contributed equally to the manuscript. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Recovering
ReactionOutcome
CC BY
33090261
20,027,693
2021-08
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Hepatotoxicity'.
First Case Report of Fulminant Hepatitis After Laparoscopic Sleeve Gastrectomy Associated with Concomitant Maximal Therapeutic Dose of Acetaminophen Use, Protein Calorie Malnutrition, and Vitamins A and D, Selenium, and Glutathione Deficiencies. Nonalcoholic fatty liver disease (NAFLD) is increasingly being linked to obesity. Although laparoscopic sleeve gastrectomy (LSG) is effective for weight loss that can ultimately resolve NAFLD, an initial transient deterioration of liver functions could be observed during the first few months post-operatively, after which a subsequent improvement of the liver functions might occur. Rapid weight loss, nutritional deficiencies, and protein malnutrition can all contribute to hepatic dysfunction and can affect the metabolism of medications such as acetaminophen leading to more insult to a compromised liver. We report acute liver failure after LSG associated with protein calorie malnutrition, multiple nutritional deficiencies in addition to concomitant use of therapeutic doses of acetaminophen. Treatment with N-acetylcysteine, and replacement of deficient multivitamins and trace elements resulted in significant improvement in liver functions. Background Nonalcoholic fatty liver disease (NAFLD) is due to an increased deposition of triglycerides into the hepatocytes to around 5% of liver weight [1]. NAFLD ranges from simple steatosis (relatively benign) to severe nonalcoholic steatohepatitis (NASH, can lead to cirrhosis and hepatocellular carcinoma) [1]. NAFLD is increasingly observed as a complication of obesity and also as part of the metabolic syndrome. Laparoscopic sleeve gastrectomy (LSG) is common and effective for weight loss, acting through restrictive and hormonal mechanisms. Although such weight loss can ultimately slow/stop the progression of or resolve the NAFLD [2], however, various extents of hepatic dysfunction could be encountered post bariatric surgery (BS) due to a range of factors. For instance, after BS, an initial transient deterioration of liver functions to the extent of possible liver failure is observed during the first few months especially after Roux-en-Y gastric bypass, after which a subsequent improvement of the liver functions might occur [3]. Such deterioration could be multifactorial: rapid weight loss leading to enhanced lipolysis and increased release of endogenous free fatty acids from adipose deposits which may in turn increase the risk of liver fibrosis [2]; changes in gut microbiota may contribute to hepatic dysfunction; protein calorie malnutrition/starvation leads to autophagy resulting in liver cell necrosis; and dehydration results in poor blood supply to the liver [4, 5]. In addition, as part of the nutritional deficiencies encountered after BS, low vitamin D levels can be associated with severe histologic changes in NAFLD; low vitamin A was linked to progression of NAFLD; and selenium deficiency is associated with decreased protection against oxidative stress [6–9]. Amidst such collective insults, common medications could prove an additional burden on an already compromised liver [10]. We report a fulminant hepatic failure 2 months after LSG accompanying protein calorie malnutrition and vitamins A, D and selenium deficiencies, associated with concomitant maximal therapeutic dose of acetaminophen use and possible glutathione deficiency. Case Report A 26-year-old Qatari female presented to the emergency room at our institution (Hamad Medical Corporation, largest tertiary care center in Qatar) on 1 Nov. 2019, complaining of a 3-week nausea, repeated vomiting and severe upper abdominal pain radiating to the back, with no aggravating factors and minimally relieved by paracetamol. She had normal bowel habits but decreased frequency and amount of urine. The patient reported fatigue and bilateral numbness episodes of the fingertips that resolved spontaneously, but no fever, skin lesions, or skin color change. She had no other sensory complaints weakness, dizziness, or visual complaints. Past history was remarkable for obesity class 3 (BMI 40 kg/m2) and benign intracranial hypertension controlled with acetazolamide. Two months earlier, weighing 95 kg (BMI 40 kg/m2), she underwent LSG that reduced her weight to 79 kg, and was off acetazolamide. Post-LSG, she tolerated pureed but not soft diet because of nausea. She denied blood transfusions, recent travel, smoking or alcohol consumption, contact with sick persons, but reported nonadherence to the prescribed multivitamins and high protein supplements. Upon examination, she was vitally stable, oriented, with clear chest and normal cardiovascular and central nervous system, normal bowel sounds, but right upper abdominal quadrant tenderness. Liver enzymes were mildly deranged (Fig. 1 B1), US of the liver showed fatty parenchymal echogenicity and calcular cholecystitis (Fig. 1 B2).Fig. 1 Timeline and sequence of events. LSG Laparoscopic sleeve gastrectomy, US ultrasound. Reference values: WBC white cell count (4–10 × 103/uL), Hct hematocrit (36–46%), MCV Mean corpuscular volume (83–101 fL), Hb hemoglobin (12–15 g/dl), Plt platelet (150- 400 × 103/uL), Alk Phos alkaline phosphatase (35–104 U/L), ALT alanine aminotransferase (0–33 U/L), AST aspartate aminotransferase (0–32 U/L), total bilirubin (0-21umol/L), total protein (66-87 g/L), albumin (35–52 g/L), PT (9.7–11.8 s), APTT (24.6–31.2 s), INR 1, amylase (13–60 U/L), lipase (13–53 U/L), ammonia (11–51 umol/L), folate (10.4–42.4 nmol/L), iron profile: iron (6–35 umol/L), TIBC total iron binding capacity (45–80 umol/L), Fe % saturation (15–45%), transferrin (2–3.6 g/L), ferritin (12–114 μg/L), vitamin A (1.05–2.09 umol/L), vitamin B12 (133–675 pmol/L), zinc (10.1–16.8 umol/L ), selenium (70–150 ng/ml), vitamin D (35–88 ng/mL), copper (11.8–22.8 umol/L), K potassium (3.5–5.1 mmol/L), Ca calcium (2.2–2.5 mmol/L), Mg magnesium (0.66–1.07 mmol/L), P phosphorus (0.81–1.45 mmol/L), ANA antinuclear antibody, ANCA antineutrophil cytoplasmic antibodies The acute surgery team admitted her and commenced treatment (Fig.1C). Four days later, with more abdominal pain, unimproved nausea and vomiting, and acute liver failure (ALF) with grade II hepatic encephalopathy (Fig.1 D1), she was transferred to intensive care unit (Fig.1 D2), where nutritional, hepatoxic, viral serologies, auto-immune profiles work ups were undertaken, as well as CT and US of the abdomen (Fig. 1 D3, D4, and D5). The gastroenterology team considered liver transplant; however, her liver function gradually improved over the following 2 days, and she was extubated (Fig. 1E). She was transferred to the ward on 12 November (Fig.1F), followed up by a multidisciplinary team, was gradually tolerating soft mechanical diet, remained asymptomatic and showed significant improvement in liver function. She was discharged on 19 November 2019 (Fig. 1G). Discussion We report a patient with history of mild derangement in liver function on the day prior to her LSG. Two months post-LSG, she presented at our hospital (index admission) and was diagnosed as calculus cholecystitis. At this stage, she had mildly deranged liver and was admitted by the general surgery team and started on treatment. Our first encounter with her was on day 5 of this index admission, where she was drowsy with lethargy, moderate confusion, asterixis, and severe transaminitis. Hence, we diagnosed acute liver failure. Liver failure post bariatric surgery has been described after some of the “older” procedures, e.g., jejunoileal bypass and biliopancreatic diversion, but is rare in modern BS, e.g., LSG [11]. Nevertheless, ALF is encountered post BS due to a range of factors [5], as depicted in Fig. 2 for our patient.Fig. 2 Multiple concomitant risk factors for liver toxicity after bariatric surgery. Capital letters within brackets in the boxes refer to the evidence available to bariatric team for suspicion of the given cause (from Fig. 1) to the liver toxicity encountered in our patient. GSH glutathione, NAFLD nonalcoholic liver disease, NASH nonalcoholic steatohepatitis. * indicates speculated factors, with no evidence available to the bariatric team for its direct effects on hepatic dysfunction/toxicity in the current patient As a procedure, LSG seems not directly implicated in ALF. On the contrary, BS generally has positive impacts on liver enzymes and histology. Particularly, LSG could be beneficial in decreasing the systemic oxidative stress observed with obesity, with positive prognosis for NAFLD/NASH patients [12, 13]. A meta-analysis (15 studies, 766 liver biopsies) observed significant improvements in the NAFLD components, namely, liver steatosis, steatohepatitis, and fibrosis in 91.6%, 81.3%, and 65.5% of patients, respectively, and complete resolution among 69.5% of patients for nonalcoholic steatohepatitis after BS [14]. As for rapid weight loss after LSG, within the previous 7 weeks pre-admission, our patient lost 16 kg (Fig. 1. B1), amounting to 2.2 kg per week. Rapid weight loss (> 1.6 kg per week) may increase visceral free fatty acids and proinflammatory cytokines, which increase the risk of liver fibrosis [2]. Likewise, rapid weight loss may also precipitate mild lobular hepatitis [15]. In addition, the associated protein malnutrition and resultant rapid mobilization of intra−/extrahepatic fat stores during weight loss could aggravate preexistent liver steatosis in these patients [15]. Collectively, such mechanisms can lead to ALF as observed in the current case. Related to the protein malnutrition and rapid weight loss is the alteration of micronutrient absorption after BS. Our patient had vitamins A, D and selenium deficiencies (Fig.1 D3). In terms of vitamin A, declining circulating and hepatic retinol levels are associated with progression of NAFLD to NASH, cirrhosis, and cancer [8]. Likewise, low vitamin D was associated with more severe histologic changes in NAFLD [6]. High selenium levels were associated with increased prevalence of NAFLD [16]; and conversely, selenium deficiency was associated with increased liver damage in experimental NAFLD models, where zinc and selenium co-supplementation improved the serum biochemical parameters such as liver enzymes and lipid profile with reductions in fat granule accumulation in the liver and liver size [9]. Such conjoint micronutrient deficiencies could have contributed to the patient’s acute liver failure. As for exogenous insults, some medications commonly prescribed to bariatric patients can contribute to liver deterioration. Following most BS types, patients avoid nonsteroidal anti-inflammatory drugs given the increased risk of such drugs for gastrointestinal ulceration. Thus, acetaminophen is among the remaining non-narcotic analgesics suitable for such patients [7]. However, baseline nutritional status might predispose to more severe acetaminophen liver injury [10]. Among patients with acetaminophen-associated ALF, those with prior BS had higher INR, lower serum albumin, and a trend toward a higher coma grade [7]. In agreement, our patient was on maximal therapeutic dose of acetaminophen and had high INR (6), low albumin (27 g/L), and moderate cognitive impairment. Her blood acetaminophen level was low (Fig.1 D4); hence, therapeutic doses could prove toxic to a liver already compromised by a range of factors as outlined above [12]. As regards to GSH, it plays a key role in the protection of the liver by detoxification of both endogenous and exogenous toxic metabolites [17]. With acetaminophen, hepatotoxicity is not caused by the drug itself, but by its reactive intermediate N-acetyl-p-benzoquinone imine (NAPQI) [18]. It is unclear whether post-BS patients have lower intrahepatic GSH stores, which limits NAPQI detoxification, causing liver injury, as seen in patients with malnutrition or chronic alcohol abuse [19]. N-acetylcysteine (NAC) replenishes intracellular GSH levels [20], thus enhancing the liver to remove toxic metabolites. There are no facilities to measure GSH at our institution, and we cannot confirm GSH deficiency in our patient; however, administration of NAC resulted in positive response with significant liver function improvements within a few days. In terms of preexisting health issues, we encountered a hospitalized, rather sick patient. Hence, we did not undertake invasive liver biopsy and cannot confirm whether she had NAFLD at this stage. Nevertheless, we are also unable to confirm that she did not have preexisting NAFLD for three reasons: her hospital records showed mild derangement of liver function on the day prior to her LSG; NAFLD can still be observed despite normal serum liver enzyme levels [21]; and index admission US abdomen showed normal liver size but fatty parenchymal echogenicity. Conclusion Despite the many positive outcomes of LSG, hepatic dysfunction, fulminant hepatitis, and liver failure can sometimes be observed due to complex interlacing factors. Rapid weight loss, protein malnutrition, macro-/micronutritional deficiencies, and the generated oxidative stress could collectively contribute to hepatic dysfunction and negatively affect the metabolism of common medications such as therapeutic acetaminophen doses leading to additional insult to a liver already compromised by NAFLD/NASH as outlined. Close monitoring and multidisciplinary follow-up of patients after bariatric surgery are recommended for prevention, early detection, and management of such conditions, along with the cautious use of acetaminophen in vulnerable patients. Open Access funding provided by the Qatar National Library. The authors appreciate the willingness of the patient to agree to this case report. Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflicts of interest. Ethical Approval All procedures performed in the study involving human participant were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This case report has been approved by the Medical Research center (IRB) (Approval # MRC-04-20-689). Informed Consent Due to the COVID-19 pandemic, written informed consent was not possible as it was deemed unethical that the patient travels to the hospital to sign the consent. Hence, informed verbal consent was obtained over the telephone from the patient after a through explanation of the fact that her case will be published in a scientific journal without breaking her confidentiality or disclosing her identity and she agreed to do so. The informed verbal consent over the telephone was witnessed by another co-author. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ACETAMINOPHEN, AMPICILLIN SODIUM\SULBACTAM SODIUM, DEXAMETHASONE, ONDANSETRON
DrugsGivenReaction
CC BY
33090351
18,518,353
2021-02
What was the administration route of drug 'AMPICILLIN SODIUM\SULBACTAM SODIUM'?
First Case Report of Fulminant Hepatitis After Laparoscopic Sleeve Gastrectomy Associated with Concomitant Maximal Therapeutic Dose of Acetaminophen Use, Protein Calorie Malnutrition, and Vitamins A and D, Selenium, and Glutathione Deficiencies. Nonalcoholic fatty liver disease (NAFLD) is increasingly being linked to obesity. Although laparoscopic sleeve gastrectomy (LSG) is effective for weight loss that can ultimately resolve NAFLD, an initial transient deterioration of liver functions could be observed during the first few months post-operatively, after which a subsequent improvement of the liver functions might occur. Rapid weight loss, nutritional deficiencies, and protein malnutrition can all contribute to hepatic dysfunction and can affect the metabolism of medications such as acetaminophen leading to more insult to a compromised liver. We report acute liver failure after LSG associated with protein calorie malnutrition, multiple nutritional deficiencies in addition to concomitant use of therapeutic doses of acetaminophen. Treatment with N-acetylcysteine, and replacement of deficient multivitamins and trace elements resulted in significant improvement in liver functions. Background Nonalcoholic fatty liver disease (NAFLD) is due to an increased deposition of triglycerides into the hepatocytes to around 5% of liver weight [1]. NAFLD ranges from simple steatosis (relatively benign) to severe nonalcoholic steatohepatitis (NASH, can lead to cirrhosis and hepatocellular carcinoma) [1]. NAFLD is increasingly observed as a complication of obesity and also as part of the metabolic syndrome. Laparoscopic sleeve gastrectomy (LSG) is common and effective for weight loss, acting through restrictive and hormonal mechanisms. Although such weight loss can ultimately slow/stop the progression of or resolve the NAFLD [2], however, various extents of hepatic dysfunction could be encountered post bariatric surgery (BS) due to a range of factors. For instance, after BS, an initial transient deterioration of liver functions to the extent of possible liver failure is observed during the first few months especially after Roux-en-Y gastric bypass, after which a subsequent improvement of the liver functions might occur [3]. Such deterioration could be multifactorial: rapid weight loss leading to enhanced lipolysis and increased release of endogenous free fatty acids from adipose deposits which may in turn increase the risk of liver fibrosis [2]; changes in gut microbiota may contribute to hepatic dysfunction; protein calorie malnutrition/starvation leads to autophagy resulting in liver cell necrosis; and dehydration results in poor blood supply to the liver [4, 5]. In addition, as part of the nutritional deficiencies encountered after BS, low vitamin D levels can be associated with severe histologic changes in NAFLD; low vitamin A was linked to progression of NAFLD; and selenium deficiency is associated with decreased protection against oxidative stress [6–9]. Amidst such collective insults, common medications could prove an additional burden on an already compromised liver [10]. We report a fulminant hepatic failure 2 months after LSG accompanying protein calorie malnutrition and vitamins A, D and selenium deficiencies, associated with concomitant maximal therapeutic dose of acetaminophen use and possible glutathione deficiency. Case Report A 26-year-old Qatari female presented to the emergency room at our institution (Hamad Medical Corporation, largest tertiary care center in Qatar) on 1 Nov. 2019, complaining of a 3-week nausea, repeated vomiting and severe upper abdominal pain radiating to the back, with no aggravating factors and minimally relieved by paracetamol. She had normal bowel habits but decreased frequency and amount of urine. The patient reported fatigue and bilateral numbness episodes of the fingertips that resolved spontaneously, but no fever, skin lesions, or skin color change. She had no other sensory complaints weakness, dizziness, or visual complaints. Past history was remarkable for obesity class 3 (BMI 40 kg/m2) and benign intracranial hypertension controlled with acetazolamide. Two months earlier, weighing 95 kg (BMI 40 kg/m2), she underwent LSG that reduced her weight to 79 kg, and was off acetazolamide. Post-LSG, she tolerated pureed but not soft diet because of nausea. She denied blood transfusions, recent travel, smoking or alcohol consumption, contact with sick persons, but reported nonadherence to the prescribed multivitamins and high protein supplements. Upon examination, she was vitally stable, oriented, with clear chest and normal cardiovascular and central nervous system, normal bowel sounds, but right upper abdominal quadrant tenderness. Liver enzymes were mildly deranged (Fig. 1 B1), US of the liver showed fatty parenchymal echogenicity and calcular cholecystitis (Fig. 1 B2).Fig. 1 Timeline and sequence of events. LSG Laparoscopic sleeve gastrectomy, US ultrasound. Reference values: WBC white cell count (4–10 × 103/uL), Hct hematocrit (36–46%), MCV Mean corpuscular volume (83–101 fL), Hb hemoglobin (12–15 g/dl), Plt platelet (150- 400 × 103/uL), Alk Phos alkaline phosphatase (35–104 U/L), ALT alanine aminotransferase (0–33 U/L), AST aspartate aminotransferase (0–32 U/L), total bilirubin (0-21umol/L), total protein (66-87 g/L), albumin (35–52 g/L), PT (9.7–11.8 s), APTT (24.6–31.2 s), INR 1, amylase (13–60 U/L), lipase (13–53 U/L), ammonia (11–51 umol/L), folate (10.4–42.4 nmol/L), iron profile: iron (6–35 umol/L), TIBC total iron binding capacity (45–80 umol/L), Fe % saturation (15–45%), transferrin (2–3.6 g/L), ferritin (12–114 μg/L), vitamin A (1.05–2.09 umol/L), vitamin B12 (133–675 pmol/L), zinc (10.1–16.8 umol/L ), selenium (70–150 ng/ml), vitamin D (35–88 ng/mL), copper (11.8–22.8 umol/L), K potassium (3.5–5.1 mmol/L), Ca calcium (2.2–2.5 mmol/L), Mg magnesium (0.66–1.07 mmol/L), P phosphorus (0.81–1.45 mmol/L), ANA antinuclear antibody, ANCA antineutrophil cytoplasmic antibodies The acute surgery team admitted her and commenced treatment (Fig.1C). Four days later, with more abdominal pain, unimproved nausea and vomiting, and acute liver failure (ALF) with grade II hepatic encephalopathy (Fig.1 D1), she was transferred to intensive care unit (Fig.1 D2), where nutritional, hepatoxic, viral serologies, auto-immune profiles work ups were undertaken, as well as CT and US of the abdomen (Fig. 1 D3, D4, and D5). The gastroenterology team considered liver transplant; however, her liver function gradually improved over the following 2 days, and she was extubated (Fig. 1E). She was transferred to the ward on 12 November (Fig.1F), followed up by a multidisciplinary team, was gradually tolerating soft mechanical diet, remained asymptomatic and showed significant improvement in liver function. She was discharged on 19 November 2019 (Fig. 1G). Discussion We report a patient with history of mild derangement in liver function on the day prior to her LSG. Two months post-LSG, she presented at our hospital (index admission) and was diagnosed as calculus cholecystitis. At this stage, she had mildly deranged liver and was admitted by the general surgery team and started on treatment. Our first encounter with her was on day 5 of this index admission, where she was drowsy with lethargy, moderate confusion, asterixis, and severe transaminitis. Hence, we diagnosed acute liver failure. Liver failure post bariatric surgery has been described after some of the “older” procedures, e.g., jejunoileal bypass and biliopancreatic diversion, but is rare in modern BS, e.g., LSG [11]. Nevertheless, ALF is encountered post BS due to a range of factors [5], as depicted in Fig. 2 for our patient.Fig. 2 Multiple concomitant risk factors for liver toxicity after bariatric surgery. Capital letters within brackets in the boxes refer to the evidence available to bariatric team for suspicion of the given cause (from Fig. 1) to the liver toxicity encountered in our patient. GSH glutathione, NAFLD nonalcoholic liver disease, NASH nonalcoholic steatohepatitis. * indicates speculated factors, with no evidence available to the bariatric team for its direct effects on hepatic dysfunction/toxicity in the current patient As a procedure, LSG seems not directly implicated in ALF. On the contrary, BS generally has positive impacts on liver enzymes and histology. Particularly, LSG could be beneficial in decreasing the systemic oxidative stress observed with obesity, with positive prognosis for NAFLD/NASH patients [12, 13]. A meta-analysis (15 studies, 766 liver biopsies) observed significant improvements in the NAFLD components, namely, liver steatosis, steatohepatitis, and fibrosis in 91.6%, 81.3%, and 65.5% of patients, respectively, and complete resolution among 69.5% of patients for nonalcoholic steatohepatitis after BS [14]. As for rapid weight loss after LSG, within the previous 7 weeks pre-admission, our patient lost 16 kg (Fig. 1. B1), amounting to 2.2 kg per week. Rapid weight loss (> 1.6 kg per week) may increase visceral free fatty acids and proinflammatory cytokines, which increase the risk of liver fibrosis [2]. Likewise, rapid weight loss may also precipitate mild lobular hepatitis [15]. In addition, the associated protein malnutrition and resultant rapid mobilization of intra−/extrahepatic fat stores during weight loss could aggravate preexistent liver steatosis in these patients [15]. Collectively, such mechanisms can lead to ALF as observed in the current case. Related to the protein malnutrition and rapid weight loss is the alteration of micronutrient absorption after BS. Our patient had vitamins A, D and selenium deficiencies (Fig.1 D3). In terms of vitamin A, declining circulating and hepatic retinol levels are associated with progression of NAFLD to NASH, cirrhosis, and cancer [8]. Likewise, low vitamin D was associated with more severe histologic changes in NAFLD [6]. High selenium levels were associated with increased prevalence of NAFLD [16]; and conversely, selenium deficiency was associated with increased liver damage in experimental NAFLD models, where zinc and selenium co-supplementation improved the serum biochemical parameters such as liver enzymes and lipid profile with reductions in fat granule accumulation in the liver and liver size [9]. Such conjoint micronutrient deficiencies could have contributed to the patient’s acute liver failure. As for exogenous insults, some medications commonly prescribed to bariatric patients can contribute to liver deterioration. Following most BS types, patients avoid nonsteroidal anti-inflammatory drugs given the increased risk of such drugs for gastrointestinal ulceration. Thus, acetaminophen is among the remaining non-narcotic analgesics suitable for such patients [7]. However, baseline nutritional status might predispose to more severe acetaminophen liver injury [10]. Among patients with acetaminophen-associated ALF, those with prior BS had higher INR, lower serum albumin, and a trend toward a higher coma grade [7]. In agreement, our patient was on maximal therapeutic dose of acetaminophen and had high INR (6), low albumin (27 g/L), and moderate cognitive impairment. Her blood acetaminophen level was low (Fig.1 D4); hence, therapeutic doses could prove toxic to a liver already compromised by a range of factors as outlined above [12]. As regards to GSH, it plays a key role in the protection of the liver by detoxification of both endogenous and exogenous toxic metabolites [17]. With acetaminophen, hepatotoxicity is not caused by the drug itself, but by its reactive intermediate N-acetyl-p-benzoquinone imine (NAPQI) [18]. It is unclear whether post-BS patients have lower intrahepatic GSH stores, which limits NAPQI detoxification, causing liver injury, as seen in patients with malnutrition or chronic alcohol abuse [19]. N-acetylcysteine (NAC) replenishes intracellular GSH levels [20], thus enhancing the liver to remove toxic metabolites. There are no facilities to measure GSH at our institution, and we cannot confirm GSH deficiency in our patient; however, administration of NAC resulted in positive response with significant liver function improvements within a few days. In terms of preexisting health issues, we encountered a hospitalized, rather sick patient. Hence, we did not undertake invasive liver biopsy and cannot confirm whether she had NAFLD at this stage. Nevertheless, we are also unable to confirm that she did not have preexisting NAFLD for three reasons: her hospital records showed mild derangement of liver function on the day prior to her LSG; NAFLD can still be observed despite normal serum liver enzyme levels [21]; and index admission US abdomen showed normal liver size but fatty parenchymal echogenicity. Conclusion Despite the many positive outcomes of LSG, hepatic dysfunction, fulminant hepatitis, and liver failure can sometimes be observed due to complex interlacing factors. Rapid weight loss, protein malnutrition, macro-/micronutritional deficiencies, and the generated oxidative stress could collectively contribute to hepatic dysfunction and negatively affect the metabolism of common medications such as therapeutic acetaminophen doses leading to additional insult to a liver already compromised by NAFLD/NASH as outlined. Close monitoring and multidisciplinary follow-up of patients after bariatric surgery are recommended for prevention, early detection, and management of such conditions, along with the cautious use of acetaminophen in vulnerable patients. Open Access funding provided by the Qatar National Library. The authors appreciate the willingness of the patient to agree to this case report. Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflicts of interest. Ethical Approval All procedures performed in the study involving human participant were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This case report has been approved by the Medical Research center (IRB) (Approval # MRC-04-20-689). Informed Consent Due to the COVID-19 pandemic, written informed consent was not possible as it was deemed unethical that the patient travels to the hospital to sign the consent. Hence, informed verbal consent was obtained over the telephone from the patient after a through explanation of the fact that her case will be published in a scientific journal without breaking her confidentiality or disclosing her identity and she agreed to do so. The informed verbal consent over the telephone was witnessed by another co-author. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33090351
18,518,353
2021-02
What was the administration route of drug 'DEXAMETHASONE'?
First Case Report of Fulminant Hepatitis After Laparoscopic Sleeve Gastrectomy Associated with Concomitant Maximal Therapeutic Dose of Acetaminophen Use, Protein Calorie Malnutrition, and Vitamins A and D, Selenium, and Glutathione Deficiencies. Nonalcoholic fatty liver disease (NAFLD) is increasingly being linked to obesity. Although laparoscopic sleeve gastrectomy (LSG) is effective for weight loss that can ultimately resolve NAFLD, an initial transient deterioration of liver functions could be observed during the first few months post-operatively, after which a subsequent improvement of the liver functions might occur. Rapid weight loss, nutritional deficiencies, and protein malnutrition can all contribute to hepatic dysfunction and can affect the metabolism of medications such as acetaminophen leading to more insult to a compromised liver. We report acute liver failure after LSG associated with protein calorie malnutrition, multiple nutritional deficiencies in addition to concomitant use of therapeutic doses of acetaminophen. Treatment with N-acetylcysteine, and replacement of deficient multivitamins and trace elements resulted in significant improvement in liver functions. Background Nonalcoholic fatty liver disease (NAFLD) is due to an increased deposition of triglycerides into the hepatocytes to around 5% of liver weight [1]. NAFLD ranges from simple steatosis (relatively benign) to severe nonalcoholic steatohepatitis (NASH, can lead to cirrhosis and hepatocellular carcinoma) [1]. NAFLD is increasingly observed as a complication of obesity and also as part of the metabolic syndrome. Laparoscopic sleeve gastrectomy (LSG) is common and effective for weight loss, acting through restrictive and hormonal mechanisms. Although such weight loss can ultimately slow/stop the progression of or resolve the NAFLD [2], however, various extents of hepatic dysfunction could be encountered post bariatric surgery (BS) due to a range of factors. For instance, after BS, an initial transient deterioration of liver functions to the extent of possible liver failure is observed during the first few months especially after Roux-en-Y gastric bypass, after which a subsequent improvement of the liver functions might occur [3]. Such deterioration could be multifactorial: rapid weight loss leading to enhanced lipolysis and increased release of endogenous free fatty acids from adipose deposits which may in turn increase the risk of liver fibrosis [2]; changes in gut microbiota may contribute to hepatic dysfunction; protein calorie malnutrition/starvation leads to autophagy resulting in liver cell necrosis; and dehydration results in poor blood supply to the liver [4, 5]. In addition, as part of the nutritional deficiencies encountered after BS, low vitamin D levels can be associated with severe histologic changes in NAFLD; low vitamin A was linked to progression of NAFLD; and selenium deficiency is associated with decreased protection against oxidative stress [6–9]. Amidst such collective insults, common medications could prove an additional burden on an already compromised liver [10]. We report a fulminant hepatic failure 2 months after LSG accompanying protein calorie malnutrition and vitamins A, D and selenium deficiencies, associated with concomitant maximal therapeutic dose of acetaminophen use and possible glutathione deficiency. Case Report A 26-year-old Qatari female presented to the emergency room at our institution (Hamad Medical Corporation, largest tertiary care center in Qatar) on 1 Nov. 2019, complaining of a 3-week nausea, repeated vomiting and severe upper abdominal pain radiating to the back, with no aggravating factors and minimally relieved by paracetamol. She had normal bowel habits but decreased frequency and amount of urine. The patient reported fatigue and bilateral numbness episodes of the fingertips that resolved spontaneously, but no fever, skin lesions, or skin color change. She had no other sensory complaints weakness, dizziness, or visual complaints. Past history was remarkable for obesity class 3 (BMI 40 kg/m2) and benign intracranial hypertension controlled with acetazolamide. Two months earlier, weighing 95 kg (BMI 40 kg/m2), she underwent LSG that reduced her weight to 79 kg, and was off acetazolamide. Post-LSG, she tolerated pureed but not soft diet because of nausea. She denied blood transfusions, recent travel, smoking or alcohol consumption, contact with sick persons, but reported nonadherence to the prescribed multivitamins and high protein supplements. Upon examination, she was vitally stable, oriented, with clear chest and normal cardiovascular and central nervous system, normal bowel sounds, but right upper abdominal quadrant tenderness. Liver enzymes were mildly deranged (Fig. 1 B1), US of the liver showed fatty parenchymal echogenicity and calcular cholecystitis (Fig. 1 B2).Fig. 1 Timeline and sequence of events. LSG Laparoscopic sleeve gastrectomy, US ultrasound. Reference values: WBC white cell count (4–10 × 103/uL), Hct hematocrit (36–46%), MCV Mean corpuscular volume (83–101 fL), Hb hemoglobin (12–15 g/dl), Plt platelet (150- 400 × 103/uL), Alk Phos alkaline phosphatase (35–104 U/L), ALT alanine aminotransferase (0–33 U/L), AST aspartate aminotransferase (0–32 U/L), total bilirubin (0-21umol/L), total protein (66-87 g/L), albumin (35–52 g/L), PT (9.7–11.8 s), APTT (24.6–31.2 s), INR 1, amylase (13–60 U/L), lipase (13–53 U/L), ammonia (11–51 umol/L), folate (10.4–42.4 nmol/L), iron profile: iron (6–35 umol/L), TIBC total iron binding capacity (45–80 umol/L), Fe % saturation (15–45%), transferrin (2–3.6 g/L), ferritin (12–114 μg/L), vitamin A (1.05–2.09 umol/L), vitamin B12 (133–675 pmol/L), zinc (10.1–16.8 umol/L ), selenium (70–150 ng/ml), vitamin D (35–88 ng/mL), copper (11.8–22.8 umol/L), K potassium (3.5–5.1 mmol/L), Ca calcium (2.2–2.5 mmol/L), Mg magnesium (0.66–1.07 mmol/L), P phosphorus (0.81–1.45 mmol/L), ANA antinuclear antibody, ANCA antineutrophil cytoplasmic antibodies The acute surgery team admitted her and commenced treatment (Fig.1C). Four days later, with more abdominal pain, unimproved nausea and vomiting, and acute liver failure (ALF) with grade II hepatic encephalopathy (Fig.1 D1), she was transferred to intensive care unit (Fig.1 D2), where nutritional, hepatoxic, viral serologies, auto-immune profiles work ups were undertaken, as well as CT and US of the abdomen (Fig. 1 D3, D4, and D5). The gastroenterology team considered liver transplant; however, her liver function gradually improved over the following 2 days, and she was extubated (Fig. 1E). She was transferred to the ward on 12 November (Fig.1F), followed up by a multidisciplinary team, was gradually tolerating soft mechanical diet, remained asymptomatic and showed significant improvement in liver function. She was discharged on 19 November 2019 (Fig. 1G). Discussion We report a patient with history of mild derangement in liver function on the day prior to her LSG. Two months post-LSG, she presented at our hospital (index admission) and was diagnosed as calculus cholecystitis. At this stage, she had mildly deranged liver and was admitted by the general surgery team and started on treatment. Our first encounter with her was on day 5 of this index admission, where she was drowsy with lethargy, moderate confusion, asterixis, and severe transaminitis. Hence, we diagnosed acute liver failure. Liver failure post bariatric surgery has been described after some of the “older” procedures, e.g., jejunoileal bypass and biliopancreatic diversion, but is rare in modern BS, e.g., LSG [11]. Nevertheless, ALF is encountered post BS due to a range of factors [5], as depicted in Fig. 2 for our patient.Fig. 2 Multiple concomitant risk factors for liver toxicity after bariatric surgery. Capital letters within brackets in the boxes refer to the evidence available to bariatric team for suspicion of the given cause (from Fig. 1) to the liver toxicity encountered in our patient. GSH glutathione, NAFLD nonalcoholic liver disease, NASH nonalcoholic steatohepatitis. * indicates speculated factors, with no evidence available to the bariatric team for its direct effects on hepatic dysfunction/toxicity in the current patient As a procedure, LSG seems not directly implicated in ALF. On the contrary, BS generally has positive impacts on liver enzymes and histology. Particularly, LSG could be beneficial in decreasing the systemic oxidative stress observed with obesity, with positive prognosis for NAFLD/NASH patients [12, 13]. A meta-analysis (15 studies, 766 liver biopsies) observed significant improvements in the NAFLD components, namely, liver steatosis, steatohepatitis, and fibrosis in 91.6%, 81.3%, and 65.5% of patients, respectively, and complete resolution among 69.5% of patients for nonalcoholic steatohepatitis after BS [14]. As for rapid weight loss after LSG, within the previous 7 weeks pre-admission, our patient lost 16 kg (Fig. 1. B1), amounting to 2.2 kg per week. Rapid weight loss (> 1.6 kg per week) may increase visceral free fatty acids and proinflammatory cytokines, which increase the risk of liver fibrosis [2]. Likewise, rapid weight loss may also precipitate mild lobular hepatitis [15]. In addition, the associated protein malnutrition and resultant rapid mobilization of intra−/extrahepatic fat stores during weight loss could aggravate preexistent liver steatosis in these patients [15]. Collectively, such mechanisms can lead to ALF as observed in the current case. Related to the protein malnutrition and rapid weight loss is the alteration of micronutrient absorption after BS. Our patient had vitamins A, D and selenium deficiencies (Fig.1 D3). In terms of vitamin A, declining circulating and hepatic retinol levels are associated with progression of NAFLD to NASH, cirrhosis, and cancer [8]. Likewise, low vitamin D was associated with more severe histologic changes in NAFLD [6]. High selenium levels were associated with increased prevalence of NAFLD [16]; and conversely, selenium deficiency was associated with increased liver damage in experimental NAFLD models, where zinc and selenium co-supplementation improved the serum biochemical parameters such as liver enzymes and lipid profile with reductions in fat granule accumulation in the liver and liver size [9]. Such conjoint micronutrient deficiencies could have contributed to the patient’s acute liver failure. As for exogenous insults, some medications commonly prescribed to bariatric patients can contribute to liver deterioration. Following most BS types, patients avoid nonsteroidal anti-inflammatory drugs given the increased risk of such drugs for gastrointestinal ulceration. Thus, acetaminophen is among the remaining non-narcotic analgesics suitable for such patients [7]. However, baseline nutritional status might predispose to more severe acetaminophen liver injury [10]. Among patients with acetaminophen-associated ALF, those with prior BS had higher INR, lower serum albumin, and a trend toward a higher coma grade [7]. In agreement, our patient was on maximal therapeutic dose of acetaminophen and had high INR (6), low albumin (27 g/L), and moderate cognitive impairment. Her blood acetaminophen level was low (Fig.1 D4); hence, therapeutic doses could prove toxic to a liver already compromised by a range of factors as outlined above [12]. As regards to GSH, it plays a key role in the protection of the liver by detoxification of both endogenous and exogenous toxic metabolites [17]. With acetaminophen, hepatotoxicity is not caused by the drug itself, but by its reactive intermediate N-acetyl-p-benzoquinone imine (NAPQI) [18]. It is unclear whether post-BS patients have lower intrahepatic GSH stores, which limits NAPQI detoxification, causing liver injury, as seen in patients with malnutrition or chronic alcohol abuse [19]. N-acetylcysteine (NAC) replenishes intracellular GSH levels [20], thus enhancing the liver to remove toxic metabolites. There are no facilities to measure GSH at our institution, and we cannot confirm GSH deficiency in our patient; however, administration of NAC resulted in positive response with significant liver function improvements within a few days. In terms of preexisting health issues, we encountered a hospitalized, rather sick patient. Hence, we did not undertake invasive liver biopsy and cannot confirm whether she had NAFLD at this stage. Nevertheless, we are also unable to confirm that she did not have preexisting NAFLD for three reasons: her hospital records showed mild derangement of liver function on the day prior to her LSG; NAFLD can still be observed despite normal serum liver enzyme levels [21]; and index admission US abdomen showed normal liver size but fatty parenchymal echogenicity. Conclusion Despite the many positive outcomes of LSG, hepatic dysfunction, fulminant hepatitis, and liver failure can sometimes be observed due to complex interlacing factors. Rapid weight loss, protein malnutrition, macro-/micronutritional deficiencies, and the generated oxidative stress could collectively contribute to hepatic dysfunction and negatively affect the metabolism of common medications such as therapeutic acetaminophen doses leading to additional insult to a liver already compromised by NAFLD/NASH as outlined. Close monitoring and multidisciplinary follow-up of patients after bariatric surgery are recommended for prevention, early detection, and management of such conditions, along with the cautious use of acetaminophen in vulnerable patients. Open Access funding provided by the Qatar National Library. The authors appreciate the willingness of the patient to agree to this case report. Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflicts of interest. Ethical Approval All procedures performed in the study involving human participant were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This case report has been approved by the Medical Research center (IRB) (Approval # MRC-04-20-689). Informed Consent Due to the COVID-19 pandemic, written informed consent was not possible as it was deemed unethical that the patient travels to the hospital to sign the consent. Hence, informed verbal consent was obtained over the telephone from the patient after a through explanation of the fact that her case will be published in a scientific journal without breaking her confidentiality or disclosing her identity and she agreed to do so. The informed verbal consent over the telephone was witnessed by another co-author. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33090351
18,525,642
2021-02
What was the administration route of drug 'ONDANSETRON'?
First Case Report of Fulminant Hepatitis After Laparoscopic Sleeve Gastrectomy Associated with Concomitant Maximal Therapeutic Dose of Acetaminophen Use, Protein Calorie Malnutrition, and Vitamins A and D, Selenium, and Glutathione Deficiencies. Nonalcoholic fatty liver disease (NAFLD) is increasingly being linked to obesity. Although laparoscopic sleeve gastrectomy (LSG) is effective for weight loss that can ultimately resolve NAFLD, an initial transient deterioration of liver functions could be observed during the first few months post-operatively, after which a subsequent improvement of the liver functions might occur. Rapid weight loss, nutritional deficiencies, and protein malnutrition can all contribute to hepatic dysfunction and can affect the metabolism of medications such as acetaminophen leading to more insult to a compromised liver. We report acute liver failure after LSG associated with protein calorie malnutrition, multiple nutritional deficiencies in addition to concomitant use of therapeutic doses of acetaminophen. Treatment with N-acetylcysteine, and replacement of deficient multivitamins and trace elements resulted in significant improvement in liver functions. Background Nonalcoholic fatty liver disease (NAFLD) is due to an increased deposition of triglycerides into the hepatocytes to around 5% of liver weight [1]. NAFLD ranges from simple steatosis (relatively benign) to severe nonalcoholic steatohepatitis (NASH, can lead to cirrhosis and hepatocellular carcinoma) [1]. NAFLD is increasingly observed as a complication of obesity and also as part of the metabolic syndrome. Laparoscopic sleeve gastrectomy (LSG) is common and effective for weight loss, acting through restrictive and hormonal mechanisms. Although such weight loss can ultimately slow/stop the progression of or resolve the NAFLD [2], however, various extents of hepatic dysfunction could be encountered post bariatric surgery (BS) due to a range of factors. For instance, after BS, an initial transient deterioration of liver functions to the extent of possible liver failure is observed during the first few months especially after Roux-en-Y gastric bypass, after which a subsequent improvement of the liver functions might occur [3]. Such deterioration could be multifactorial: rapid weight loss leading to enhanced lipolysis and increased release of endogenous free fatty acids from adipose deposits which may in turn increase the risk of liver fibrosis [2]; changes in gut microbiota may contribute to hepatic dysfunction; protein calorie malnutrition/starvation leads to autophagy resulting in liver cell necrosis; and dehydration results in poor blood supply to the liver [4, 5]. In addition, as part of the nutritional deficiencies encountered after BS, low vitamin D levels can be associated with severe histologic changes in NAFLD; low vitamin A was linked to progression of NAFLD; and selenium deficiency is associated with decreased protection against oxidative stress [6–9]. Amidst such collective insults, common medications could prove an additional burden on an already compromised liver [10]. We report a fulminant hepatic failure 2 months after LSG accompanying protein calorie malnutrition and vitamins A, D and selenium deficiencies, associated with concomitant maximal therapeutic dose of acetaminophen use and possible glutathione deficiency. Case Report A 26-year-old Qatari female presented to the emergency room at our institution (Hamad Medical Corporation, largest tertiary care center in Qatar) on 1 Nov. 2019, complaining of a 3-week nausea, repeated vomiting and severe upper abdominal pain radiating to the back, with no aggravating factors and minimally relieved by paracetamol. She had normal bowel habits but decreased frequency and amount of urine. The patient reported fatigue and bilateral numbness episodes of the fingertips that resolved spontaneously, but no fever, skin lesions, or skin color change. She had no other sensory complaints weakness, dizziness, or visual complaints. Past history was remarkable for obesity class 3 (BMI 40 kg/m2) and benign intracranial hypertension controlled with acetazolamide. Two months earlier, weighing 95 kg (BMI 40 kg/m2), she underwent LSG that reduced her weight to 79 kg, and was off acetazolamide. Post-LSG, she tolerated pureed but not soft diet because of nausea. She denied blood transfusions, recent travel, smoking or alcohol consumption, contact with sick persons, but reported nonadherence to the prescribed multivitamins and high protein supplements. Upon examination, she was vitally stable, oriented, with clear chest and normal cardiovascular and central nervous system, normal bowel sounds, but right upper abdominal quadrant tenderness. Liver enzymes were mildly deranged (Fig. 1 B1), US of the liver showed fatty parenchymal echogenicity and calcular cholecystitis (Fig. 1 B2).Fig. 1 Timeline and sequence of events. LSG Laparoscopic sleeve gastrectomy, US ultrasound. Reference values: WBC white cell count (4–10 × 103/uL), Hct hematocrit (36–46%), MCV Mean corpuscular volume (83–101 fL), Hb hemoglobin (12–15 g/dl), Plt platelet (150- 400 × 103/uL), Alk Phos alkaline phosphatase (35–104 U/L), ALT alanine aminotransferase (0–33 U/L), AST aspartate aminotransferase (0–32 U/L), total bilirubin (0-21umol/L), total protein (66-87 g/L), albumin (35–52 g/L), PT (9.7–11.8 s), APTT (24.6–31.2 s), INR 1, amylase (13–60 U/L), lipase (13–53 U/L), ammonia (11–51 umol/L), folate (10.4–42.4 nmol/L), iron profile: iron (6–35 umol/L), TIBC total iron binding capacity (45–80 umol/L), Fe % saturation (15–45%), transferrin (2–3.6 g/L), ferritin (12–114 μg/L), vitamin A (1.05–2.09 umol/L), vitamin B12 (133–675 pmol/L), zinc (10.1–16.8 umol/L ), selenium (70–150 ng/ml), vitamin D (35–88 ng/mL), copper (11.8–22.8 umol/L), K potassium (3.5–5.1 mmol/L), Ca calcium (2.2–2.5 mmol/L), Mg magnesium (0.66–1.07 mmol/L), P phosphorus (0.81–1.45 mmol/L), ANA antinuclear antibody, ANCA antineutrophil cytoplasmic antibodies The acute surgery team admitted her and commenced treatment (Fig.1C). Four days later, with more abdominal pain, unimproved nausea and vomiting, and acute liver failure (ALF) with grade II hepatic encephalopathy (Fig.1 D1), she was transferred to intensive care unit (Fig.1 D2), where nutritional, hepatoxic, viral serologies, auto-immune profiles work ups were undertaken, as well as CT and US of the abdomen (Fig. 1 D3, D4, and D5). The gastroenterology team considered liver transplant; however, her liver function gradually improved over the following 2 days, and she was extubated (Fig. 1E). She was transferred to the ward on 12 November (Fig.1F), followed up by a multidisciplinary team, was gradually tolerating soft mechanical diet, remained asymptomatic and showed significant improvement in liver function. She was discharged on 19 November 2019 (Fig. 1G). Discussion We report a patient with history of mild derangement in liver function on the day prior to her LSG. Two months post-LSG, she presented at our hospital (index admission) and was diagnosed as calculus cholecystitis. At this stage, she had mildly deranged liver and was admitted by the general surgery team and started on treatment. Our first encounter with her was on day 5 of this index admission, where she was drowsy with lethargy, moderate confusion, asterixis, and severe transaminitis. Hence, we diagnosed acute liver failure. Liver failure post bariatric surgery has been described after some of the “older” procedures, e.g., jejunoileal bypass and biliopancreatic diversion, but is rare in modern BS, e.g., LSG [11]. Nevertheless, ALF is encountered post BS due to a range of factors [5], as depicted in Fig. 2 for our patient.Fig. 2 Multiple concomitant risk factors for liver toxicity after bariatric surgery. Capital letters within brackets in the boxes refer to the evidence available to bariatric team for suspicion of the given cause (from Fig. 1) to the liver toxicity encountered in our patient. GSH glutathione, NAFLD nonalcoholic liver disease, NASH nonalcoholic steatohepatitis. * indicates speculated factors, with no evidence available to the bariatric team for its direct effects on hepatic dysfunction/toxicity in the current patient As a procedure, LSG seems not directly implicated in ALF. On the contrary, BS generally has positive impacts on liver enzymes and histology. Particularly, LSG could be beneficial in decreasing the systemic oxidative stress observed with obesity, with positive prognosis for NAFLD/NASH patients [12, 13]. A meta-analysis (15 studies, 766 liver biopsies) observed significant improvements in the NAFLD components, namely, liver steatosis, steatohepatitis, and fibrosis in 91.6%, 81.3%, and 65.5% of patients, respectively, and complete resolution among 69.5% of patients for nonalcoholic steatohepatitis after BS [14]. As for rapid weight loss after LSG, within the previous 7 weeks pre-admission, our patient lost 16 kg (Fig. 1. B1), amounting to 2.2 kg per week. Rapid weight loss (> 1.6 kg per week) may increase visceral free fatty acids and proinflammatory cytokines, which increase the risk of liver fibrosis [2]. Likewise, rapid weight loss may also precipitate mild lobular hepatitis [15]. In addition, the associated protein malnutrition and resultant rapid mobilization of intra−/extrahepatic fat stores during weight loss could aggravate preexistent liver steatosis in these patients [15]. Collectively, such mechanisms can lead to ALF as observed in the current case. Related to the protein malnutrition and rapid weight loss is the alteration of micronutrient absorption after BS. Our patient had vitamins A, D and selenium deficiencies (Fig.1 D3). In terms of vitamin A, declining circulating and hepatic retinol levels are associated with progression of NAFLD to NASH, cirrhosis, and cancer [8]. Likewise, low vitamin D was associated with more severe histologic changes in NAFLD [6]. High selenium levels were associated with increased prevalence of NAFLD [16]; and conversely, selenium deficiency was associated with increased liver damage in experimental NAFLD models, where zinc and selenium co-supplementation improved the serum biochemical parameters such as liver enzymes and lipid profile with reductions in fat granule accumulation in the liver and liver size [9]. Such conjoint micronutrient deficiencies could have contributed to the patient’s acute liver failure. As for exogenous insults, some medications commonly prescribed to bariatric patients can contribute to liver deterioration. Following most BS types, patients avoid nonsteroidal anti-inflammatory drugs given the increased risk of such drugs for gastrointestinal ulceration. Thus, acetaminophen is among the remaining non-narcotic analgesics suitable for such patients [7]. However, baseline nutritional status might predispose to more severe acetaminophen liver injury [10]. Among patients with acetaminophen-associated ALF, those with prior BS had higher INR, lower serum albumin, and a trend toward a higher coma grade [7]. In agreement, our patient was on maximal therapeutic dose of acetaminophen and had high INR (6), low albumin (27 g/L), and moderate cognitive impairment. Her blood acetaminophen level was low (Fig.1 D4); hence, therapeutic doses could prove toxic to a liver already compromised by a range of factors as outlined above [12]. As regards to GSH, it plays a key role in the protection of the liver by detoxification of both endogenous and exogenous toxic metabolites [17]. With acetaminophen, hepatotoxicity is not caused by the drug itself, but by its reactive intermediate N-acetyl-p-benzoquinone imine (NAPQI) [18]. It is unclear whether post-BS patients have lower intrahepatic GSH stores, which limits NAPQI detoxification, causing liver injury, as seen in patients with malnutrition or chronic alcohol abuse [19]. N-acetylcysteine (NAC) replenishes intracellular GSH levels [20], thus enhancing the liver to remove toxic metabolites. There are no facilities to measure GSH at our institution, and we cannot confirm GSH deficiency in our patient; however, administration of NAC resulted in positive response with significant liver function improvements within a few days. In terms of preexisting health issues, we encountered a hospitalized, rather sick patient. Hence, we did not undertake invasive liver biopsy and cannot confirm whether she had NAFLD at this stage. Nevertheless, we are also unable to confirm that she did not have preexisting NAFLD for three reasons: her hospital records showed mild derangement of liver function on the day prior to her LSG; NAFLD can still be observed despite normal serum liver enzyme levels [21]; and index admission US abdomen showed normal liver size but fatty parenchymal echogenicity. Conclusion Despite the many positive outcomes of LSG, hepatic dysfunction, fulminant hepatitis, and liver failure can sometimes be observed due to complex interlacing factors. Rapid weight loss, protein malnutrition, macro-/micronutritional deficiencies, and the generated oxidative stress could collectively contribute to hepatic dysfunction and negatively affect the metabolism of common medications such as therapeutic acetaminophen doses leading to additional insult to a liver already compromised by NAFLD/NASH as outlined. Close monitoring and multidisciplinary follow-up of patients after bariatric surgery are recommended for prevention, early detection, and management of such conditions, along with the cautious use of acetaminophen in vulnerable patients. Open Access funding provided by the Qatar National Library. The authors appreciate the willingness of the patient to agree to this case report. Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflicts of interest. Ethical Approval All procedures performed in the study involving human participant were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This case report has been approved by the Medical Research center (IRB) (Approval # MRC-04-20-689). Informed Consent Due to the COVID-19 pandemic, written informed consent was not possible as it was deemed unethical that the patient travels to the hospital to sign the consent. Hence, informed verbal consent was obtained over the telephone from the patient after a through explanation of the fact that her case will be published in a scientific journal without breaking her confidentiality or disclosing her identity and she agreed to do so. The informed verbal consent over the telephone was witnessed by another co-author. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Intravenous (not otherwise specified)
DrugAdministrationRoute
CC BY
33090351
18,518,353
2021-02
What was the dosage of drug 'ACETAMINOPHEN\HYDROCODONE'?
First Case Report of Fulminant Hepatitis After Laparoscopic Sleeve Gastrectomy Associated with Concomitant Maximal Therapeutic Dose of Acetaminophen Use, Protein Calorie Malnutrition, and Vitamins A and D, Selenium, and Glutathione Deficiencies. Nonalcoholic fatty liver disease (NAFLD) is increasingly being linked to obesity. Although laparoscopic sleeve gastrectomy (LSG) is effective for weight loss that can ultimately resolve NAFLD, an initial transient deterioration of liver functions could be observed during the first few months post-operatively, after which a subsequent improvement of the liver functions might occur. Rapid weight loss, nutritional deficiencies, and protein malnutrition can all contribute to hepatic dysfunction and can affect the metabolism of medications such as acetaminophen leading to more insult to a compromised liver. We report acute liver failure after LSG associated with protein calorie malnutrition, multiple nutritional deficiencies in addition to concomitant use of therapeutic doses of acetaminophen. Treatment with N-acetylcysteine, and replacement of deficient multivitamins and trace elements resulted in significant improvement in liver functions. Background Nonalcoholic fatty liver disease (NAFLD) is due to an increased deposition of triglycerides into the hepatocytes to around 5% of liver weight [1]. NAFLD ranges from simple steatosis (relatively benign) to severe nonalcoholic steatohepatitis (NASH, can lead to cirrhosis and hepatocellular carcinoma) [1]. NAFLD is increasingly observed as a complication of obesity and also as part of the metabolic syndrome. Laparoscopic sleeve gastrectomy (LSG) is common and effective for weight loss, acting through restrictive and hormonal mechanisms. Although such weight loss can ultimately slow/stop the progression of or resolve the NAFLD [2], however, various extents of hepatic dysfunction could be encountered post bariatric surgery (BS) due to a range of factors. For instance, after BS, an initial transient deterioration of liver functions to the extent of possible liver failure is observed during the first few months especially after Roux-en-Y gastric bypass, after which a subsequent improvement of the liver functions might occur [3]. Such deterioration could be multifactorial: rapid weight loss leading to enhanced lipolysis and increased release of endogenous free fatty acids from adipose deposits which may in turn increase the risk of liver fibrosis [2]; changes in gut microbiota may contribute to hepatic dysfunction; protein calorie malnutrition/starvation leads to autophagy resulting in liver cell necrosis; and dehydration results in poor blood supply to the liver [4, 5]. In addition, as part of the nutritional deficiencies encountered after BS, low vitamin D levels can be associated with severe histologic changes in NAFLD; low vitamin A was linked to progression of NAFLD; and selenium deficiency is associated with decreased protection against oxidative stress [6–9]. Amidst such collective insults, common medications could prove an additional burden on an already compromised liver [10]. We report a fulminant hepatic failure 2 months after LSG accompanying protein calorie malnutrition and vitamins A, D and selenium deficiencies, associated with concomitant maximal therapeutic dose of acetaminophen use and possible glutathione deficiency. Case Report A 26-year-old Qatari female presented to the emergency room at our institution (Hamad Medical Corporation, largest tertiary care center in Qatar) on 1 Nov. 2019, complaining of a 3-week nausea, repeated vomiting and severe upper abdominal pain radiating to the back, with no aggravating factors and minimally relieved by paracetamol. She had normal bowel habits but decreased frequency and amount of urine. The patient reported fatigue and bilateral numbness episodes of the fingertips that resolved spontaneously, but no fever, skin lesions, or skin color change. She had no other sensory complaints weakness, dizziness, or visual complaints. Past history was remarkable for obesity class 3 (BMI 40 kg/m2) and benign intracranial hypertension controlled with acetazolamide. Two months earlier, weighing 95 kg (BMI 40 kg/m2), she underwent LSG that reduced her weight to 79 kg, and was off acetazolamide. Post-LSG, she tolerated pureed but not soft diet because of nausea. She denied blood transfusions, recent travel, smoking or alcohol consumption, contact with sick persons, but reported nonadherence to the prescribed multivitamins and high protein supplements. Upon examination, she was vitally stable, oriented, with clear chest and normal cardiovascular and central nervous system, normal bowel sounds, but right upper abdominal quadrant tenderness. Liver enzymes were mildly deranged (Fig. 1 B1), US of the liver showed fatty parenchymal echogenicity and calcular cholecystitis (Fig. 1 B2).Fig. 1 Timeline and sequence of events. LSG Laparoscopic sleeve gastrectomy, US ultrasound. Reference values: WBC white cell count (4–10 × 103/uL), Hct hematocrit (36–46%), MCV Mean corpuscular volume (83–101 fL), Hb hemoglobin (12–15 g/dl), Plt platelet (150- 400 × 103/uL), Alk Phos alkaline phosphatase (35–104 U/L), ALT alanine aminotransferase (0–33 U/L), AST aspartate aminotransferase (0–32 U/L), total bilirubin (0-21umol/L), total protein (66-87 g/L), albumin (35–52 g/L), PT (9.7–11.8 s), APTT (24.6–31.2 s), INR 1, amylase (13–60 U/L), lipase (13–53 U/L), ammonia (11–51 umol/L), folate (10.4–42.4 nmol/L), iron profile: iron (6–35 umol/L), TIBC total iron binding capacity (45–80 umol/L), Fe % saturation (15–45%), transferrin (2–3.6 g/L), ferritin (12–114 μg/L), vitamin A (1.05–2.09 umol/L), vitamin B12 (133–675 pmol/L), zinc (10.1–16.8 umol/L ), selenium (70–150 ng/ml), vitamin D (35–88 ng/mL), copper (11.8–22.8 umol/L), K potassium (3.5–5.1 mmol/L), Ca calcium (2.2–2.5 mmol/L), Mg magnesium (0.66–1.07 mmol/L), P phosphorus (0.81–1.45 mmol/L), ANA antinuclear antibody, ANCA antineutrophil cytoplasmic antibodies The acute surgery team admitted her and commenced treatment (Fig.1C). Four days later, with more abdominal pain, unimproved nausea and vomiting, and acute liver failure (ALF) with grade II hepatic encephalopathy (Fig.1 D1), she was transferred to intensive care unit (Fig.1 D2), where nutritional, hepatoxic, viral serologies, auto-immune profiles work ups were undertaken, as well as CT and US of the abdomen (Fig. 1 D3, D4, and D5). The gastroenterology team considered liver transplant; however, her liver function gradually improved over the following 2 days, and she was extubated (Fig. 1E). She was transferred to the ward on 12 November (Fig.1F), followed up by a multidisciplinary team, was gradually tolerating soft mechanical diet, remained asymptomatic and showed significant improvement in liver function. She was discharged on 19 November 2019 (Fig. 1G). Discussion We report a patient with history of mild derangement in liver function on the day prior to her LSG. Two months post-LSG, she presented at our hospital (index admission) and was diagnosed as calculus cholecystitis. At this stage, she had mildly deranged liver and was admitted by the general surgery team and started on treatment. Our first encounter with her was on day 5 of this index admission, where she was drowsy with lethargy, moderate confusion, asterixis, and severe transaminitis. Hence, we diagnosed acute liver failure. Liver failure post bariatric surgery has been described after some of the “older” procedures, e.g., jejunoileal bypass and biliopancreatic diversion, but is rare in modern BS, e.g., LSG [11]. Nevertheless, ALF is encountered post BS due to a range of factors [5], as depicted in Fig. 2 for our patient.Fig. 2 Multiple concomitant risk factors for liver toxicity after bariatric surgery. Capital letters within brackets in the boxes refer to the evidence available to bariatric team for suspicion of the given cause (from Fig. 1) to the liver toxicity encountered in our patient. GSH glutathione, NAFLD nonalcoholic liver disease, NASH nonalcoholic steatohepatitis. * indicates speculated factors, with no evidence available to the bariatric team for its direct effects on hepatic dysfunction/toxicity in the current patient As a procedure, LSG seems not directly implicated in ALF. On the contrary, BS generally has positive impacts on liver enzymes and histology. Particularly, LSG could be beneficial in decreasing the systemic oxidative stress observed with obesity, with positive prognosis for NAFLD/NASH patients [12, 13]. A meta-analysis (15 studies, 766 liver biopsies) observed significant improvements in the NAFLD components, namely, liver steatosis, steatohepatitis, and fibrosis in 91.6%, 81.3%, and 65.5% of patients, respectively, and complete resolution among 69.5% of patients for nonalcoholic steatohepatitis after BS [14]. As for rapid weight loss after LSG, within the previous 7 weeks pre-admission, our patient lost 16 kg (Fig. 1. B1), amounting to 2.2 kg per week. Rapid weight loss (> 1.6 kg per week) may increase visceral free fatty acids and proinflammatory cytokines, which increase the risk of liver fibrosis [2]. Likewise, rapid weight loss may also precipitate mild lobular hepatitis [15]. In addition, the associated protein malnutrition and resultant rapid mobilization of intra−/extrahepatic fat stores during weight loss could aggravate preexistent liver steatosis in these patients [15]. Collectively, such mechanisms can lead to ALF as observed in the current case. Related to the protein malnutrition and rapid weight loss is the alteration of micronutrient absorption after BS. Our patient had vitamins A, D and selenium deficiencies (Fig.1 D3). In terms of vitamin A, declining circulating and hepatic retinol levels are associated with progression of NAFLD to NASH, cirrhosis, and cancer [8]. Likewise, low vitamin D was associated with more severe histologic changes in NAFLD [6]. High selenium levels were associated with increased prevalence of NAFLD [16]; and conversely, selenium deficiency was associated with increased liver damage in experimental NAFLD models, where zinc and selenium co-supplementation improved the serum biochemical parameters such as liver enzymes and lipid profile with reductions in fat granule accumulation in the liver and liver size [9]. Such conjoint micronutrient deficiencies could have contributed to the patient’s acute liver failure. As for exogenous insults, some medications commonly prescribed to bariatric patients can contribute to liver deterioration. Following most BS types, patients avoid nonsteroidal anti-inflammatory drugs given the increased risk of such drugs for gastrointestinal ulceration. Thus, acetaminophen is among the remaining non-narcotic analgesics suitable for such patients [7]. However, baseline nutritional status might predispose to more severe acetaminophen liver injury [10]. Among patients with acetaminophen-associated ALF, those with prior BS had higher INR, lower serum albumin, and a trend toward a higher coma grade [7]. In agreement, our patient was on maximal therapeutic dose of acetaminophen and had high INR (6), low albumin (27 g/L), and moderate cognitive impairment. Her blood acetaminophen level was low (Fig.1 D4); hence, therapeutic doses could prove toxic to a liver already compromised by a range of factors as outlined above [12]. As regards to GSH, it plays a key role in the protection of the liver by detoxification of both endogenous and exogenous toxic metabolites [17]. With acetaminophen, hepatotoxicity is not caused by the drug itself, but by its reactive intermediate N-acetyl-p-benzoquinone imine (NAPQI) [18]. It is unclear whether post-BS patients have lower intrahepatic GSH stores, which limits NAPQI detoxification, causing liver injury, as seen in patients with malnutrition or chronic alcohol abuse [19]. N-acetylcysteine (NAC) replenishes intracellular GSH levels [20], thus enhancing the liver to remove toxic metabolites. There are no facilities to measure GSH at our institution, and we cannot confirm GSH deficiency in our patient; however, administration of NAC resulted in positive response with significant liver function improvements within a few days. In terms of preexisting health issues, we encountered a hospitalized, rather sick patient. Hence, we did not undertake invasive liver biopsy and cannot confirm whether she had NAFLD at this stage. Nevertheless, we are also unable to confirm that she did not have preexisting NAFLD for three reasons: her hospital records showed mild derangement of liver function on the day prior to her LSG; NAFLD can still be observed despite normal serum liver enzyme levels [21]; and index admission US abdomen showed normal liver size but fatty parenchymal echogenicity. Conclusion Despite the many positive outcomes of LSG, hepatic dysfunction, fulminant hepatitis, and liver failure can sometimes be observed due to complex interlacing factors. Rapid weight loss, protein malnutrition, macro-/micronutritional deficiencies, and the generated oxidative stress could collectively contribute to hepatic dysfunction and negatively affect the metabolism of common medications such as therapeutic acetaminophen doses leading to additional insult to a liver already compromised by NAFLD/NASH as outlined. Close monitoring and multidisciplinary follow-up of patients after bariatric surgery are recommended for prevention, early detection, and management of such conditions, along with the cautious use of acetaminophen in vulnerable patients. Open Access funding provided by the Qatar National Library. The authors appreciate the willingness of the patient to agree to this case report. Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflicts of interest. Ethical Approval All procedures performed in the study involving human participant were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This case report has been approved by the Medical Research center (IRB) (Approval # MRC-04-20-689). Informed Consent Due to the COVID-19 pandemic, written informed consent was not possible as it was deemed unethical that the patient travels to the hospital to sign the consent. Hence, informed verbal consent was obtained over the telephone from the patient after a through explanation of the fact that her case will be published in a scientific journal without breaking her confidentiality or disclosing her identity and she agreed to do so. The informed verbal consent over the telephone was witnessed by another co-author. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
1 GRAM
DrugDosageText
CC BY
33090351
18,525,642
2021-02
What was the dosage of drug 'ONDANSETRON'?
First Case Report of Fulminant Hepatitis After Laparoscopic Sleeve Gastrectomy Associated with Concomitant Maximal Therapeutic Dose of Acetaminophen Use, Protein Calorie Malnutrition, and Vitamins A and D, Selenium, and Glutathione Deficiencies. Nonalcoholic fatty liver disease (NAFLD) is increasingly being linked to obesity. Although laparoscopic sleeve gastrectomy (LSG) is effective for weight loss that can ultimately resolve NAFLD, an initial transient deterioration of liver functions could be observed during the first few months post-operatively, after which a subsequent improvement of the liver functions might occur. Rapid weight loss, nutritional deficiencies, and protein malnutrition can all contribute to hepatic dysfunction and can affect the metabolism of medications such as acetaminophen leading to more insult to a compromised liver. We report acute liver failure after LSG associated with protein calorie malnutrition, multiple nutritional deficiencies in addition to concomitant use of therapeutic doses of acetaminophen. Treatment with N-acetylcysteine, and replacement of deficient multivitamins and trace elements resulted in significant improvement in liver functions. Background Nonalcoholic fatty liver disease (NAFLD) is due to an increased deposition of triglycerides into the hepatocytes to around 5% of liver weight [1]. NAFLD ranges from simple steatosis (relatively benign) to severe nonalcoholic steatohepatitis (NASH, can lead to cirrhosis and hepatocellular carcinoma) [1]. NAFLD is increasingly observed as a complication of obesity and also as part of the metabolic syndrome. Laparoscopic sleeve gastrectomy (LSG) is common and effective for weight loss, acting through restrictive and hormonal mechanisms. Although such weight loss can ultimately slow/stop the progression of or resolve the NAFLD [2], however, various extents of hepatic dysfunction could be encountered post bariatric surgery (BS) due to a range of factors. For instance, after BS, an initial transient deterioration of liver functions to the extent of possible liver failure is observed during the first few months especially after Roux-en-Y gastric bypass, after which a subsequent improvement of the liver functions might occur [3]. Such deterioration could be multifactorial: rapid weight loss leading to enhanced lipolysis and increased release of endogenous free fatty acids from adipose deposits which may in turn increase the risk of liver fibrosis [2]; changes in gut microbiota may contribute to hepatic dysfunction; protein calorie malnutrition/starvation leads to autophagy resulting in liver cell necrosis; and dehydration results in poor blood supply to the liver [4, 5]. In addition, as part of the nutritional deficiencies encountered after BS, low vitamin D levels can be associated with severe histologic changes in NAFLD; low vitamin A was linked to progression of NAFLD; and selenium deficiency is associated with decreased protection against oxidative stress [6–9]. Amidst such collective insults, common medications could prove an additional burden on an already compromised liver [10]. We report a fulminant hepatic failure 2 months after LSG accompanying protein calorie malnutrition and vitamins A, D and selenium deficiencies, associated with concomitant maximal therapeutic dose of acetaminophen use and possible glutathione deficiency. Case Report A 26-year-old Qatari female presented to the emergency room at our institution (Hamad Medical Corporation, largest tertiary care center in Qatar) on 1 Nov. 2019, complaining of a 3-week nausea, repeated vomiting and severe upper abdominal pain radiating to the back, with no aggravating factors and minimally relieved by paracetamol. She had normal bowel habits but decreased frequency and amount of urine. The patient reported fatigue and bilateral numbness episodes of the fingertips that resolved spontaneously, but no fever, skin lesions, or skin color change. She had no other sensory complaints weakness, dizziness, or visual complaints. Past history was remarkable for obesity class 3 (BMI 40 kg/m2) and benign intracranial hypertension controlled with acetazolamide. Two months earlier, weighing 95 kg (BMI 40 kg/m2), she underwent LSG that reduced her weight to 79 kg, and was off acetazolamide. Post-LSG, she tolerated pureed but not soft diet because of nausea. She denied blood transfusions, recent travel, smoking or alcohol consumption, contact with sick persons, but reported nonadherence to the prescribed multivitamins and high protein supplements. Upon examination, she was vitally stable, oriented, with clear chest and normal cardiovascular and central nervous system, normal bowel sounds, but right upper abdominal quadrant tenderness. Liver enzymes were mildly deranged (Fig. 1 B1), US of the liver showed fatty parenchymal echogenicity and calcular cholecystitis (Fig. 1 B2).Fig. 1 Timeline and sequence of events. LSG Laparoscopic sleeve gastrectomy, US ultrasound. Reference values: WBC white cell count (4–10 × 103/uL), Hct hematocrit (36–46%), MCV Mean corpuscular volume (83–101 fL), Hb hemoglobin (12–15 g/dl), Plt platelet (150- 400 × 103/uL), Alk Phos alkaline phosphatase (35–104 U/L), ALT alanine aminotransferase (0–33 U/L), AST aspartate aminotransferase (0–32 U/L), total bilirubin (0-21umol/L), total protein (66-87 g/L), albumin (35–52 g/L), PT (9.7–11.8 s), APTT (24.6–31.2 s), INR 1, amylase (13–60 U/L), lipase (13–53 U/L), ammonia (11–51 umol/L), folate (10.4–42.4 nmol/L), iron profile: iron (6–35 umol/L), TIBC total iron binding capacity (45–80 umol/L), Fe % saturation (15–45%), transferrin (2–3.6 g/L), ferritin (12–114 μg/L), vitamin A (1.05–2.09 umol/L), vitamin B12 (133–675 pmol/L), zinc (10.1–16.8 umol/L ), selenium (70–150 ng/ml), vitamin D (35–88 ng/mL), copper (11.8–22.8 umol/L), K potassium (3.5–5.1 mmol/L), Ca calcium (2.2–2.5 mmol/L), Mg magnesium (0.66–1.07 mmol/L), P phosphorus (0.81–1.45 mmol/L), ANA antinuclear antibody, ANCA antineutrophil cytoplasmic antibodies The acute surgery team admitted her and commenced treatment (Fig.1C). Four days later, with more abdominal pain, unimproved nausea and vomiting, and acute liver failure (ALF) with grade II hepatic encephalopathy (Fig.1 D1), she was transferred to intensive care unit (Fig.1 D2), where nutritional, hepatoxic, viral serologies, auto-immune profiles work ups were undertaken, as well as CT and US of the abdomen (Fig. 1 D3, D4, and D5). The gastroenterology team considered liver transplant; however, her liver function gradually improved over the following 2 days, and she was extubated (Fig. 1E). She was transferred to the ward on 12 November (Fig.1F), followed up by a multidisciplinary team, was gradually tolerating soft mechanical diet, remained asymptomatic and showed significant improvement in liver function. She was discharged on 19 November 2019 (Fig. 1G). Discussion We report a patient with history of mild derangement in liver function on the day prior to her LSG. Two months post-LSG, she presented at our hospital (index admission) and was diagnosed as calculus cholecystitis. At this stage, she had mildly deranged liver and was admitted by the general surgery team and started on treatment. Our first encounter with her was on day 5 of this index admission, where she was drowsy with lethargy, moderate confusion, asterixis, and severe transaminitis. Hence, we diagnosed acute liver failure. Liver failure post bariatric surgery has been described after some of the “older” procedures, e.g., jejunoileal bypass and biliopancreatic diversion, but is rare in modern BS, e.g., LSG [11]. Nevertheless, ALF is encountered post BS due to a range of factors [5], as depicted in Fig. 2 for our patient.Fig. 2 Multiple concomitant risk factors for liver toxicity after bariatric surgery. Capital letters within brackets in the boxes refer to the evidence available to bariatric team for suspicion of the given cause (from Fig. 1) to the liver toxicity encountered in our patient. GSH glutathione, NAFLD nonalcoholic liver disease, NASH nonalcoholic steatohepatitis. * indicates speculated factors, with no evidence available to the bariatric team for its direct effects on hepatic dysfunction/toxicity in the current patient As a procedure, LSG seems not directly implicated in ALF. On the contrary, BS generally has positive impacts on liver enzymes and histology. Particularly, LSG could be beneficial in decreasing the systemic oxidative stress observed with obesity, with positive prognosis for NAFLD/NASH patients [12, 13]. A meta-analysis (15 studies, 766 liver biopsies) observed significant improvements in the NAFLD components, namely, liver steatosis, steatohepatitis, and fibrosis in 91.6%, 81.3%, and 65.5% of patients, respectively, and complete resolution among 69.5% of patients for nonalcoholic steatohepatitis after BS [14]. As for rapid weight loss after LSG, within the previous 7 weeks pre-admission, our patient lost 16 kg (Fig. 1. B1), amounting to 2.2 kg per week. Rapid weight loss (> 1.6 kg per week) may increase visceral free fatty acids and proinflammatory cytokines, which increase the risk of liver fibrosis [2]. Likewise, rapid weight loss may also precipitate mild lobular hepatitis [15]. In addition, the associated protein malnutrition and resultant rapid mobilization of intra−/extrahepatic fat stores during weight loss could aggravate preexistent liver steatosis in these patients [15]. Collectively, such mechanisms can lead to ALF as observed in the current case. Related to the protein malnutrition and rapid weight loss is the alteration of micronutrient absorption after BS. Our patient had vitamins A, D and selenium deficiencies (Fig.1 D3). In terms of vitamin A, declining circulating and hepatic retinol levels are associated with progression of NAFLD to NASH, cirrhosis, and cancer [8]. Likewise, low vitamin D was associated with more severe histologic changes in NAFLD [6]. High selenium levels were associated with increased prevalence of NAFLD [16]; and conversely, selenium deficiency was associated with increased liver damage in experimental NAFLD models, where zinc and selenium co-supplementation improved the serum biochemical parameters such as liver enzymes and lipid profile with reductions in fat granule accumulation in the liver and liver size [9]. Such conjoint micronutrient deficiencies could have contributed to the patient’s acute liver failure. As for exogenous insults, some medications commonly prescribed to bariatric patients can contribute to liver deterioration. Following most BS types, patients avoid nonsteroidal anti-inflammatory drugs given the increased risk of such drugs for gastrointestinal ulceration. Thus, acetaminophen is among the remaining non-narcotic analgesics suitable for such patients [7]. However, baseline nutritional status might predispose to more severe acetaminophen liver injury [10]. Among patients with acetaminophen-associated ALF, those with prior BS had higher INR, lower serum albumin, and a trend toward a higher coma grade [7]. In agreement, our patient was on maximal therapeutic dose of acetaminophen and had high INR (6), low albumin (27 g/L), and moderate cognitive impairment. Her blood acetaminophen level was low (Fig.1 D4); hence, therapeutic doses could prove toxic to a liver already compromised by a range of factors as outlined above [12]. As regards to GSH, it plays a key role in the protection of the liver by detoxification of both endogenous and exogenous toxic metabolites [17]. With acetaminophen, hepatotoxicity is not caused by the drug itself, but by its reactive intermediate N-acetyl-p-benzoquinone imine (NAPQI) [18]. It is unclear whether post-BS patients have lower intrahepatic GSH stores, which limits NAPQI detoxification, causing liver injury, as seen in patients with malnutrition or chronic alcohol abuse [19]. N-acetylcysteine (NAC) replenishes intracellular GSH levels [20], thus enhancing the liver to remove toxic metabolites. There are no facilities to measure GSH at our institution, and we cannot confirm GSH deficiency in our patient; however, administration of NAC resulted in positive response with significant liver function improvements within a few days. In terms of preexisting health issues, we encountered a hospitalized, rather sick patient. Hence, we did not undertake invasive liver biopsy and cannot confirm whether she had NAFLD at this stage. Nevertheless, we are also unable to confirm that she did not have preexisting NAFLD for three reasons: her hospital records showed mild derangement of liver function on the day prior to her LSG; NAFLD can still be observed despite normal serum liver enzyme levels [21]; and index admission US abdomen showed normal liver size but fatty parenchymal echogenicity. Conclusion Despite the many positive outcomes of LSG, hepatic dysfunction, fulminant hepatitis, and liver failure can sometimes be observed due to complex interlacing factors. Rapid weight loss, protein malnutrition, macro-/micronutritional deficiencies, and the generated oxidative stress could collectively contribute to hepatic dysfunction and negatively affect the metabolism of common medications such as therapeutic acetaminophen doses leading to additional insult to a liver already compromised by NAFLD/NASH as outlined. Close monitoring and multidisciplinary follow-up of patients after bariatric surgery are recommended for prevention, early detection, and management of such conditions, along with the cautious use of acetaminophen in vulnerable patients. Open Access funding provided by the Qatar National Library. The authors appreciate the willingness of the patient to agree to this case report. Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflicts of interest. Ethical Approval All procedures performed in the study involving human participant were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This case report has been approved by the Medical Research center (IRB) (Approval # MRC-04-20-689). Informed Consent Due to the COVID-19 pandemic, written informed consent was not possible as it was deemed unethical that the patient travels to the hospital to sign the consent. Hence, informed verbal consent was obtained over the telephone from the patient after a through explanation of the fact that her case will be published in a scientific journal without breaking her confidentiality or disclosing her identity and she agreed to do so. The informed verbal consent over the telephone was witnessed by another co-author. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
8 mg (milligrams).
DrugDosage
CC BY
33090351
18,518,353
2021-02
What was the outcome of reaction 'Hepatotoxicity'?
First Case Report of Fulminant Hepatitis After Laparoscopic Sleeve Gastrectomy Associated with Concomitant Maximal Therapeutic Dose of Acetaminophen Use, Protein Calorie Malnutrition, and Vitamins A and D, Selenium, and Glutathione Deficiencies. Nonalcoholic fatty liver disease (NAFLD) is increasingly being linked to obesity. Although laparoscopic sleeve gastrectomy (LSG) is effective for weight loss that can ultimately resolve NAFLD, an initial transient deterioration of liver functions could be observed during the first few months post-operatively, after which a subsequent improvement of the liver functions might occur. Rapid weight loss, nutritional deficiencies, and protein malnutrition can all contribute to hepatic dysfunction and can affect the metabolism of medications such as acetaminophen leading to more insult to a compromised liver. We report acute liver failure after LSG associated with protein calorie malnutrition, multiple nutritional deficiencies in addition to concomitant use of therapeutic doses of acetaminophen. Treatment with N-acetylcysteine, and replacement of deficient multivitamins and trace elements resulted in significant improvement in liver functions. Background Nonalcoholic fatty liver disease (NAFLD) is due to an increased deposition of triglycerides into the hepatocytes to around 5% of liver weight [1]. NAFLD ranges from simple steatosis (relatively benign) to severe nonalcoholic steatohepatitis (NASH, can lead to cirrhosis and hepatocellular carcinoma) [1]. NAFLD is increasingly observed as a complication of obesity and also as part of the metabolic syndrome. Laparoscopic sleeve gastrectomy (LSG) is common and effective for weight loss, acting through restrictive and hormonal mechanisms. Although such weight loss can ultimately slow/stop the progression of or resolve the NAFLD [2], however, various extents of hepatic dysfunction could be encountered post bariatric surgery (BS) due to a range of factors. For instance, after BS, an initial transient deterioration of liver functions to the extent of possible liver failure is observed during the first few months especially after Roux-en-Y gastric bypass, after which a subsequent improvement of the liver functions might occur [3]. Such deterioration could be multifactorial: rapid weight loss leading to enhanced lipolysis and increased release of endogenous free fatty acids from adipose deposits which may in turn increase the risk of liver fibrosis [2]; changes in gut microbiota may contribute to hepatic dysfunction; protein calorie malnutrition/starvation leads to autophagy resulting in liver cell necrosis; and dehydration results in poor blood supply to the liver [4, 5]. In addition, as part of the nutritional deficiencies encountered after BS, low vitamin D levels can be associated with severe histologic changes in NAFLD; low vitamin A was linked to progression of NAFLD; and selenium deficiency is associated with decreased protection against oxidative stress [6–9]. Amidst such collective insults, common medications could prove an additional burden on an already compromised liver [10]. We report a fulminant hepatic failure 2 months after LSG accompanying protein calorie malnutrition and vitamins A, D and selenium deficiencies, associated with concomitant maximal therapeutic dose of acetaminophen use and possible glutathione deficiency. Case Report A 26-year-old Qatari female presented to the emergency room at our institution (Hamad Medical Corporation, largest tertiary care center in Qatar) on 1 Nov. 2019, complaining of a 3-week nausea, repeated vomiting and severe upper abdominal pain radiating to the back, with no aggravating factors and minimally relieved by paracetamol. She had normal bowel habits but decreased frequency and amount of urine. The patient reported fatigue and bilateral numbness episodes of the fingertips that resolved spontaneously, but no fever, skin lesions, or skin color change. She had no other sensory complaints weakness, dizziness, or visual complaints. Past history was remarkable for obesity class 3 (BMI 40 kg/m2) and benign intracranial hypertension controlled with acetazolamide. Two months earlier, weighing 95 kg (BMI 40 kg/m2), she underwent LSG that reduced her weight to 79 kg, and was off acetazolamide. Post-LSG, she tolerated pureed but not soft diet because of nausea. She denied blood transfusions, recent travel, smoking or alcohol consumption, contact with sick persons, but reported nonadherence to the prescribed multivitamins and high protein supplements. Upon examination, she was vitally stable, oriented, with clear chest and normal cardiovascular and central nervous system, normal bowel sounds, but right upper abdominal quadrant tenderness. Liver enzymes were mildly deranged (Fig. 1 B1), US of the liver showed fatty parenchymal echogenicity and calcular cholecystitis (Fig. 1 B2).Fig. 1 Timeline and sequence of events. LSG Laparoscopic sleeve gastrectomy, US ultrasound. Reference values: WBC white cell count (4–10 × 103/uL), Hct hematocrit (36–46%), MCV Mean corpuscular volume (83–101 fL), Hb hemoglobin (12–15 g/dl), Plt platelet (150- 400 × 103/uL), Alk Phos alkaline phosphatase (35–104 U/L), ALT alanine aminotransferase (0–33 U/L), AST aspartate aminotransferase (0–32 U/L), total bilirubin (0-21umol/L), total protein (66-87 g/L), albumin (35–52 g/L), PT (9.7–11.8 s), APTT (24.6–31.2 s), INR 1, amylase (13–60 U/L), lipase (13–53 U/L), ammonia (11–51 umol/L), folate (10.4–42.4 nmol/L), iron profile: iron (6–35 umol/L), TIBC total iron binding capacity (45–80 umol/L), Fe % saturation (15–45%), transferrin (2–3.6 g/L), ferritin (12–114 μg/L), vitamin A (1.05–2.09 umol/L), vitamin B12 (133–675 pmol/L), zinc (10.1–16.8 umol/L ), selenium (70–150 ng/ml), vitamin D (35–88 ng/mL), copper (11.8–22.8 umol/L), K potassium (3.5–5.1 mmol/L), Ca calcium (2.2–2.5 mmol/L), Mg magnesium (0.66–1.07 mmol/L), P phosphorus (0.81–1.45 mmol/L), ANA antinuclear antibody, ANCA antineutrophil cytoplasmic antibodies The acute surgery team admitted her and commenced treatment (Fig.1C). Four days later, with more abdominal pain, unimproved nausea and vomiting, and acute liver failure (ALF) with grade II hepatic encephalopathy (Fig.1 D1), she was transferred to intensive care unit (Fig.1 D2), where nutritional, hepatoxic, viral serologies, auto-immune profiles work ups were undertaken, as well as CT and US of the abdomen (Fig. 1 D3, D4, and D5). The gastroenterology team considered liver transplant; however, her liver function gradually improved over the following 2 days, and she was extubated (Fig. 1E). She was transferred to the ward on 12 November (Fig.1F), followed up by a multidisciplinary team, was gradually tolerating soft mechanical diet, remained asymptomatic and showed significant improvement in liver function. She was discharged on 19 November 2019 (Fig. 1G). Discussion We report a patient with history of mild derangement in liver function on the day prior to her LSG. Two months post-LSG, she presented at our hospital (index admission) and was diagnosed as calculus cholecystitis. At this stage, she had mildly deranged liver and was admitted by the general surgery team and started on treatment. Our first encounter with her was on day 5 of this index admission, where she was drowsy with lethargy, moderate confusion, asterixis, and severe transaminitis. Hence, we diagnosed acute liver failure. Liver failure post bariatric surgery has been described after some of the “older” procedures, e.g., jejunoileal bypass and biliopancreatic diversion, but is rare in modern BS, e.g., LSG [11]. Nevertheless, ALF is encountered post BS due to a range of factors [5], as depicted in Fig. 2 for our patient.Fig. 2 Multiple concomitant risk factors for liver toxicity after bariatric surgery. Capital letters within brackets in the boxes refer to the evidence available to bariatric team for suspicion of the given cause (from Fig. 1) to the liver toxicity encountered in our patient. GSH glutathione, NAFLD nonalcoholic liver disease, NASH nonalcoholic steatohepatitis. * indicates speculated factors, with no evidence available to the bariatric team for its direct effects on hepatic dysfunction/toxicity in the current patient As a procedure, LSG seems not directly implicated in ALF. On the contrary, BS generally has positive impacts on liver enzymes and histology. Particularly, LSG could be beneficial in decreasing the systemic oxidative stress observed with obesity, with positive prognosis for NAFLD/NASH patients [12, 13]. A meta-analysis (15 studies, 766 liver biopsies) observed significant improvements in the NAFLD components, namely, liver steatosis, steatohepatitis, and fibrosis in 91.6%, 81.3%, and 65.5% of patients, respectively, and complete resolution among 69.5% of patients for nonalcoholic steatohepatitis after BS [14]. As for rapid weight loss after LSG, within the previous 7 weeks pre-admission, our patient lost 16 kg (Fig. 1. B1), amounting to 2.2 kg per week. Rapid weight loss (> 1.6 kg per week) may increase visceral free fatty acids and proinflammatory cytokines, which increase the risk of liver fibrosis [2]. Likewise, rapid weight loss may also precipitate mild lobular hepatitis [15]. In addition, the associated protein malnutrition and resultant rapid mobilization of intra−/extrahepatic fat stores during weight loss could aggravate preexistent liver steatosis in these patients [15]. Collectively, such mechanisms can lead to ALF as observed in the current case. Related to the protein malnutrition and rapid weight loss is the alteration of micronutrient absorption after BS. Our patient had vitamins A, D and selenium deficiencies (Fig.1 D3). In terms of vitamin A, declining circulating and hepatic retinol levels are associated with progression of NAFLD to NASH, cirrhosis, and cancer [8]. Likewise, low vitamin D was associated with more severe histologic changes in NAFLD [6]. High selenium levels were associated with increased prevalence of NAFLD [16]; and conversely, selenium deficiency was associated with increased liver damage in experimental NAFLD models, where zinc and selenium co-supplementation improved the serum biochemical parameters such as liver enzymes and lipid profile with reductions in fat granule accumulation in the liver and liver size [9]. Such conjoint micronutrient deficiencies could have contributed to the patient’s acute liver failure. As for exogenous insults, some medications commonly prescribed to bariatric patients can contribute to liver deterioration. Following most BS types, patients avoid nonsteroidal anti-inflammatory drugs given the increased risk of such drugs for gastrointestinal ulceration. Thus, acetaminophen is among the remaining non-narcotic analgesics suitable for such patients [7]. However, baseline nutritional status might predispose to more severe acetaminophen liver injury [10]. Among patients with acetaminophen-associated ALF, those with prior BS had higher INR, lower serum albumin, and a trend toward a higher coma grade [7]. In agreement, our patient was on maximal therapeutic dose of acetaminophen and had high INR (6), low albumin (27 g/L), and moderate cognitive impairment. Her blood acetaminophen level was low (Fig.1 D4); hence, therapeutic doses could prove toxic to a liver already compromised by a range of factors as outlined above [12]. As regards to GSH, it plays a key role in the protection of the liver by detoxification of both endogenous and exogenous toxic metabolites [17]. With acetaminophen, hepatotoxicity is not caused by the drug itself, but by its reactive intermediate N-acetyl-p-benzoquinone imine (NAPQI) [18]. It is unclear whether post-BS patients have lower intrahepatic GSH stores, which limits NAPQI detoxification, causing liver injury, as seen in patients with malnutrition or chronic alcohol abuse [19]. N-acetylcysteine (NAC) replenishes intracellular GSH levels [20], thus enhancing the liver to remove toxic metabolites. There are no facilities to measure GSH at our institution, and we cannot confirm GSH deficiency in our patient; however, administration of NAC resulted in positive response with significant liver function improvements within a few days. In terms of preexisting health issues, we encountered a hospitalized, rather sick patient. Hence, we did not undertake invasive liver biopsy and cannot confirm whether she had NAFLD at this stage. Nevertheless, we are also unable to confirm that she did not have preexisting NAFLD for three reasons: her hospital records showed mild derangement of liver function on the day prior to her LSG; NAFLD can still be observed despite normal serum liver enzyme levels [21]; and index admission US abdomen showed normal liver size but fatty parenchymal echogenicity. Conclusion Despite the many positive outcomes of LSG, hepatic dysfunction, fulminant hepatitis, and liver failure can sometimes be observed due to complex interlacing factors. Rapid weight loss, protein malnutrition, macro-/micronutritional deficiencies, and the generated oxidative stress could collectively contribute to hepatic dysfunction and negatively affect the metabolism of common medications such as therapeutic acetaminophen doses leading to additional insult to a liver already compromised by NAFLD/NASH as outlined. Close monitoring and multidisciplinary follow-up of patients after bariatric surgery are recommended for prevention, early detection, and management of such conditions, along with the cautious use of acetaminophen in vulnerable patients. Open Access funding provided by the Qatar National Library. The authors appreciate the willingness of the patient to agree to this case report. Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflicts of interest. Ethical Approval All procedures performed in the study involving human participant were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. This case report has been approved by the Medical Research center (IRB) (Approval # MRC-04-20-689). Informed Consent Due to the COVID-19 pandemic, written informed consent was not possible as it was deemed unethical that the patient travels to the hospital to sign the consent. Hence, informed verbal consent was obtained over the telephone from the patient after a through explanation of the fact that her case will be published in a scientific journal without breaking her confidentiality or disclosing her identity and she agreed to do so. The informed verbal consent over the telephone was witnessed by another co-author. Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Recovering
ReactionOutcome
CC BY
33090351
18,518,353
2021-02
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Cytokine release syndrome'.
Filgrastim associations with CAR T-cell therapy. Little is known about the benefits and risks of myeloid growth factor administration after chimeric antigen receptor (CAR) T-cell therapy for diffuse large B-cell lymphoma (DLBCL). We present a retrospective analysis among 22 relapsed/refractory DLBCL patients who received CAR T-cell therapy with axicabtagene ciloleucel. Filgrastim was administered by physician discretion to seven patients (31.8%), and the median duration of neutropenia after lymphodepleting therapy was significantly shorter for those patients who received filgrastim (5 vs 15 days, P = .016). Five patients (22.7%) developed infection in the 30 days post-CAR T-cell therapy with three patients being Grade 3 or higher. There was no difference in the incidence and severity of infection based on filgrastim use (P = .274, P = .138). Among the seven patients that received filgrastim, six patients (85.7%) and four patients (57.1%) had evidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), respectively. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) and 7 patients (46.7%) had evidence of CRS and ICANS, respectively. There was no significant difference in the incidence of developing CRS or ICANS between the group of patients that received filgrastim and those that did not (P = .193, P = .647). However, there was a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042). Filgrastim administration after CAR T-cell therapy may lead to an increase in severity of CRS without decreasing infection rates. Abbreviations ANCabsolute neutrophil count ASTCTAmerican Society of Transplantation and Cellular Therapy CARchimeric antigen receptor CTCAECommon Terminology Criteria for Adverse Events DLBCLdiffuse large B‐cell lymphoma G‐CSFgranulocyte colony‐stimulating factor GM‐CSFgranulocyte‐macrophage colony‐stimulating factor ICANSimmune effector cell‐associated neurotoxicity syndrome IFNinterferon ILinterleukin IQRinterquartile range RICErituximab/ifosfamide/carboplatin/etoposide 1 INTRODUCTION While chimeric antigen receptor (CAR) T‐cell therapy is an exciting advancement in the treatment of patients with relapsed/refractory diffuse large B‐cell lymphoma (DLBCL), its application is limited by associated toxicities, including cytokine release syndrome (CRS), neurotoxicity and severe cytopenias, with 78% of patients developing Grade 3 or higher neutropenia in the ZUMA‐1 trial. 1 Recombinant granulocyte colony‐stimulating factor (G‐CSF) has been widely used to shorten the duration of neutropenia and risk of infection in other settings, such as neutropenic complications after conditioning for hematopoietic cell transplant. 2 However, in CAR T‐cell therapy, myeloid growth factors have the potential to increase the incidence and/or severity of CRS and immune effector cell‐associated neurotoxicity syndrome (ICANS) by promotion of proinflammatory cytokine secretion from monocytes and macrophages. 3 , 4 , 5 Preclinically, monocytes and macrophages are the main source of interleukin (IL)‐1 and IL‐6 during CRS. 4 , 5 Surprisingly, CRS severity is mediated not by CAR T‐cell‐derived cytokines, but by those of monocytes and macrophages. 4 , 5 Therefore, we sought to determine if a relationship exists between filgrastim administration, which stimulates myelopoiesis and enhances granulocyte function, and CRS. There is currently a lack of evidence to guide clinicians on the benefits and risks of recombinant myeloid growth factor use in CAR T‐cell treatment. 2 MATERIALS AND METHODS Between March 2018 and May 2019, we reviewed 22 patients with DLBCL treated with axicabtagene ciloleucel with or without concurrent use of filgrastim. Prior to CAR T‐cell infusion, all patients received standard lymphodepleting therapy with fludarabine 30 mg/m2/day and cyclophosphamide 500 mg/m2/day on days −5 through −3, except for one patient that received reduced doses for chronic kidney disease Stage IV (fludarabine 15 mg/m2/day and cyclophosphamide 375 mg/m2/day). Prophylactic tocilizumab 8 mg/kg was given to all patients at 36 hours after CAR T‐cell infusion, with additional doses of tocilizumab and/or steroids given for evidence of CRS/ICANS based on the American Society for Transplantation and Cellular Therapy (ASTCT) consensus grading system. 6 Filgrastim was administered at physician discretion after CAR T‐cell infusion at a weight‐based dose of either 300 or 480 mcg, and cumulative filgrastim dose was recorded within the first 30 days. Toxicity after CAR T‐cell therapy was also assessed up to 30 days postinfusion. Any patient with neutropenic fever was treated with broad‐spectrum antibiotics; no prophylactic antibiotics were given. Documented infections were graded according to Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. 7 Antibiotics were also administered for any confirmed infections. Chi‐squared test or Cochran‐Armitage test were performed to examine association between cohort characteristics and the study variable of administration with or without filgrastim. 3 RESULTS Baseline patient characteristics are displayed in Table 1. The median age at the time of CAR T‐cell therapy was 65.0 years (interquartile range [IQR], 57.0‐68.8). The majority (14 patients, 63.6%) had high‐grade DLBCL, while 5 patients had DLBCL not otherwise specified, 2 patients had transformed follicular lymphoma and 1 patient had Richter's transformation. Seven patients (31.8%) had refractory DLBCL, while 15 patients (68.2%) had relapsed DLBCL with a median number of relapses prior to CAR T‐cell therapy of 1.0 (IQR, 0.0‐2.0). Thirteen patients (59.1%) had received greater than or equal to three prior therapies. Only one patient (4.6%) had received autologous hematopoietic cell transplant prior to CAR T‐cell therapy. Ten patients (45.5%) received bridging therapy for high disease burden before CAR T‐cell therapy, which included rituximab/gemcitabine‐oxaliplatin (7 patients), cytarabine/thiotepa (1 patient), rituximab/dexamethasone/cytarabine/cisplatin (1 patient), rituximab/ifosfamide/carboplatin/etoposide (RICE, 1 patient) and rituximab/cyclophosphamide/dexamethasone (1 patient). The number of cycles of bridging therapy ranged from 1 to 3 with a median of 2. TABLE 1 Baseline patient and disease characteristics All patients (N = 22) Age at time of CAR T‐cell therapy (median, IQR) 65.0 (57.0, 68.8) ≥65 12 (54.6%) <65 10 (45.5%) Gender Male 12 (54.6%) Female 10 (45.5%) Ethnicity Caucasian 12 (54.6%) Hispanic 7 (31.8%) Asian 0 (0%) African American 1 (4.6%) Other 2 (9.1%) Disease type DLBCL NOS 5 (22.7%) Transformed follicular 2 (9.1%) Richter's transformation 1 (4.6%) High‐grade DLBCL 14 (63.6%) Cell of origin GCB 10 (45.5%) Non‐GCB 10 (45.5%) Unspecified 2 (9.1%) Relapse or Refractory Relapse 15 (68.2%) Refractory 7 (31.8%) Number of prior therapies <3 9 (40.9%) ≥3 13 (59.1%) ASCT prior to CAR T‐cell therapy 1 (4.6%) ECOG at time of CAR T‐cell therapy 0‐1 21 (95.5%) 2‐4 1 (4.6%) Bridging therapy given before CAR T‐cell therapy Yes 10 (45.5%) No 12 (54.6%) Abbreviations: ASCT, autologous stem cell transplant; CAR, chimeric antigen receptor; DLBCL, diffuse large B‐cell lymphoma; ECOG, Eastern Cooperative Oncology Group; GCB, germinal center B‐cell; NOS, not otherwise specified. Seven of the 22 patients (31.8%) received filgrastim by physician discretion at a target dose of 5 mcg/kg/day with 3 patients receiving 300 mcg/day and 4 patients receiving 480 mcg/day. The median start day of filgrastim was 2 days post‐CAR T‐cell therapy (range Day −2 to Day +7), and the median number of filgrastim doses was 6 (range, 3‐18). The median duration of neutropenia after CAR T‐cell therapy was 5 days (IQR, 4.5‐8.5) for patients who received filgrastim compared to 15 days (IQR, 8.0‐30.0) for patients who did not receive filgrastim (P = .016; Table 2). Fourteen patients (63.6%) developed neutropenic fever after CAR T‐cell therapy, 6 of whom received filgrastim and 8 of whom had not (P = .193; Table 2). Five patients (22.7%) developed an infection in the 30 days post‐CAR T‐cell therapy including Clostridium difficile colitis (one patient, Grade 3), Enterococcus faecalis bacteremia (one patient, Grade 4), pneumonia (one patient, Grade 3) and Herpes simplex virus (two patients, Grades 1 and 2). Among the seven patients that received filgrastim, three patients (42.9%) developed an infection and two patients (28.6%) developed a Grade 3 or greater infection. Among the 15 patients that did not receive filgrastim, 2 patients (13.2%) developed an infection and 1 patient (6.7%) developed a Grade 3 or greater infection. There was no difference in the incidence and severity of infection between patients who received filgrastim and those that did not (P = .274, P = .138; Table 2; Figure 1). TABLE 2 CAR T‐cell associated toxicity All patients (N = 22) G‐CSF administered (N = 7) G‐CSF not administered (N = 15) P value Median duration of neutropenia (days) (IQR) 10 (6.0, 25.8) 5 (4.5, 8.5) 15 (8.0, 30.0) .016 Febrile neutropenia No 8 (36.4%) 1 (14.3%) 7 (46.7%) .193 Yes 14 (63.6%) 6 (85.7%) 8 (53.5%) New infection .274 None 17 (77.3%) 4 (57.1%) 13 (86.7%) Any grade 5 (22.7%) 3 (42.9%) 2 (13.2%) CRS .193 None 8 (36.4%) 1 (14.3%) 7 (46.7%) Any grade 14 (63.6%) 6 (85.7%) 8 (53.3%) ICANS .647 None 11 (50.0%) 3 (42.9%) 8 (53.3%) Any grade 11 (50.0%) 4 (57.1%) 7 (46.7%) Steroids given .648 No 9 (40.9%) 2 (28.6%) 7 (46.7%) Yes 13 (59.1%) 5 (71.4%) 8 (53.3%) More than one dose of tocilizumab given .074 No 10 (45.5%) 1 (14.3%) 9 (60.0%) Yes 12 (54.6%) 6 (85.7%) 6 (40.0%) Abbreviations: CAR, chimeric antigen receptor; CRS, cytokine release syndrome; G‐CSF, granulocyte colony‐stimulating factor; ICANS, immune effector cell‐associated neurotoxicity syndrome. FIGURE 1 Severity of chimeric antigen receptor (CAR) T‐cell associated toxicities based on granulocyte colony‐stimulating factor (G‐CSF) use. ICANS, immune effector cell‐associated neurotoxicity syndrome [Color figure can be viewed at wileyonlinelibrary.com] CRS was noted in 14 patients overall (63.6%), and 4 patients (18.2%) had Grade 3 or higher CRS. Among the seven patients that received filgrastim, six patients (85.7%) had evidence of CRS and three patients (42.9%) had Grade 3 or higher CRS. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) had evidence of CRS and 1 patient (6.7%) had evidence of Grade 3 or higher CRS. ICANS was noted in 11 patients overall (50.0%), and 9 patients (40.9%) had Grade 3 or higher ICANS. Among the seven patients that received filgrastim, four patients (57.1%) had evidence of ICANS and three patients (42.9%) had evidence of Grade 3 or higher ICANS. Among the 15 patients that did not receive filgrastim, 7 patients (46.7%) had evidence of ICANS and 6 patients (40.0%) had Grade 3 or higher ICANS. There was no significant difference in the incidence of developing CRS (any grade) or ICANS (any grade) between the group of patients that received filgrastim and those that did not (P = .193, P = .647; Table 2). There was, however, a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042), but no increase in the severity of ICANS based on filgrastim use (P = .660; Figure 1). Thirteen patients (59.1%) received corticosteroids after CAR T‐cell treatment at a median cumulative dosage of 666.7 mg prednisone equivalents (IQR, 445.0‐933.3), with the majority (9 patients, 69.2%) receiving steroids in the first 5 days post‐CAR T‐cell infusion. Half of the patient cohort (12 patients, 54.6%) required at least one dose of tocilizumab in addition to the scheduled prophylactic dose. Among the seven patients that received filgrastim, 5 patients (71.4%) received corticosteroids and 6 patients (85.7%) received more than one dose of tocilizumab. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) received corticosteroids and 6 patients (40.0%) received more than one dose of tocilizumab. There was no association between filgrastim use and steroid use or administration of additional doses of tocilizumab (P = .648, P = .074). 4 DISCUSSION At our institution, filgrastim was administered after CAR T‐cell therapy at physician discretion with less than half of our patient cohort receiving filgrastim (seven patients, 31.8%). Importantly, although the median duration of neutropenia was significantly lower for patients who received filgrastim, there was no difference in the development or severity of infections (P = .274, P = .138). Instead, while patients given filgrastim were not more likely to develop CRS (P = .193), the severity of CRS was higher in those that received filgrastim (P = .042), as has been suggested by preclinical data modeling myeloid growth factors in combination with CAR T‐cells. 4 , 5 The development of CRS is related to activation of in vivo T‐cell expansion and the production of T‐cell effector cytokines, including IL‐6, IL‐10 and interferon (IFN)‐γ, 8 which are also downstream products of myeloid cells stimulated by filgrastim. One preclinical study showed that higher levels of murine G‐CSF correlated strongly with CRS severity and survival. 5 Elevated systemic levels of IL‐6 in particular have been associated with severe CRS, and IL‐6 receptor blockade with the monoclonal antibody tocilizumab is an important agent used to reduce CRS toxicity. 9 , 10 Of all the cytokines analyzed in the ZUMA‐2 study, only peak levels of granzyme B and granulocyte‐macrophage colony stimulating factor (GM‐CSF, a closely related protein to G‐CSF) were associated with severe CRS and severe ICANS. 11 CAR T‐cell associated neurotoxicity has been associated with elevations in similar inflammatory markers to CRS (IL‐6, IL‐10, IFNγ), in addition to higher serum levels of G‐CSF and GM‐CSF. 12 , 13 Mouse models of CRS and ICANS have suggested that the main driver of CAR T‐cell neurotoxicity is actually IL‐1 secretion from activated macrophages. 4 , 5 Unlike its efficacy in CRS, tocilizumab did not protect mice from lethal neurotoxicity, a finding analogous in humans; however the IL‐1 receptor antagonist anakinra abolished both CRS and neurotoxicity. 4 , 12 Interestingly, in addition to its stimulatory effects on granulocytes, filgrastim has immunomodulatory effects on other immune cells and can inhibit inflammatory cytokine production within monocytes or macrophages. 14 This may account for why we found no association with filgrastim and the development or severity of neurotoxicity (P = .647, P = .660). GM‐CSF, which induces both granulocytes and macrophages, may be more closely tied to neurotoxic effects and, in fact, GM‐CSF inhibition has been shown to reduce neuroinflammation and prevent CRS. 15 The guidelines on use of filgrastim and other growth factors with CAR T‐cell therapy are not standardized. While some recommend administration of myeloid growth factors once absolute neutrophil count (ANC) decreases to <500/μL and to continue until ANC increases to ≥1500/μL, 16 others recommend administration of filgrastim only if patients develop Grade 1 CRS at the same time as neutropenia or in the setting of neutropenic fever. 17 The use of myeloid growth factors across institutions employing CAR T‐cell therapy is also mixed. In an electronic survey on the current administrative, logistic and toxicity management practices of CAR T‐cell therapy across the United States by the ASTCT, out of 28 respondents, 46% used growth factor if allowed by product labeling, 29% never administered growth factor, 14% determined the use of growth factor on a patient‐specific basis and 11% administered growth factor to all patients. 18 Although our study is limited by its small sample size and retrospective nature, we suggest that myeloid growth factor administration be used with caution in patients undergoing CAR T‐cell therapy. We found an association between filgrastim use and CRS severity, suggesting that increasing the number of patients in the study would strengthen the association. However, the effect size of the association was small. Further studies are required to determine the safety of myeloid growth factors after CAR T‐cell therapy. CONFLICT OF INTEREST The authors declared no potential conflicts of interest. ETHICS STATEMENT This study was approved by the University of California Los Angeles Institutional Review Board. Informed consent was not required for this study. ACKNOWLEDGMENT This work was supported by National Institutes of Health Grant Number 1K08CA245483‐01. Myung Shin Sim was supported by NIH National Center for Advancing Translational Sciences (Grant 5UL1TR001881‐05). DATA AVAILABILITY STATEMENT Data are available on request from the authors.
AXICABTAGENE CILOLEUCEL, CYCLOPHOSPHAMIDE, FILGRASTIM, FLUDARABINE PHOSPHATE, TOCILIZUMAB
DrugsGivenReaction
CC BY-NC
33091961
18,449,659
2021-03-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Diffuse large B-cell lymphoma'.
Filgrastim associations with CAR T-cell therapy. Little is known about the benefits and risks of myeloid growth factor administration after chimeric antigen receptor (CAR) T-cell therapy for diffuse large B-cell lymphoma (DLBCL). We present a retrospective analysis among 22 relapsed/refractory DLBCL patients who received CAR T-cell therapy with axicabtagene ciloleucel. Filgrastim was administered by physician discretion to seven patients (31.8%), and the median duration of neutropenia after lymphodepleting therapy was significantly shorter for those patients who received filgrastim (5 vs 15 days, P = .016). Five patients (22.7%) developed infection in the 30 days post-CAR T-cell therapy with three patients being Grade 3 or higher. There was no difference in the incidence and severity of infection based on filgrastim use (P = .274, P = .138). Among the seven patients that received filgrastim, six patients (85.7%) and four patients (57.1%) had evidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), respectively. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) and 7 patients (46.7%) had evidence of CRS and ICANS, respectively. There was no significant difference in the incidence of developing CRS or ICANS between the group of patients that received filgrastim and those that did not (P = .193, P = .647). However, there was a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042). Filgrastim administration after CAR T-cell therapy may lead to an increase in severity of CRS without decreasing infection rates. Abbreviations ANCabsolute neutrophil count ASTCTAmerican Society of Transplantation and Cellular Therapy CARchimeric antigen receptor CTCAECommon Terminology Criteria for Adverse Events DLBCLdiffuse large B‐cell lymphoma G‐CSFgranulocyte colony‐stimulating factor GM‐CSFgranulocyte‐macrophage colony‐stimulating factor ICANSimmune effector cell‐associated neurotoxicity syndrome IFNinterferon ILinterleukin IQRinterquartile range RICErituximab/ifosfamide/carboplatin/etoposide 1 INTRODUCTION While chimeric antigen receptor (CAR) T‐cell therapy is an exciting advancement in the treatment of patients with relapsed/refractory diffuse large B‐cell lymphoma (DLBCL), its application is limited by associated toxicities, including cytokine release syndrome (CRS), neurotoxicity and severe cytopenias, with 78% of patients developing Grade 3 or higher neutropenia in the ZUMA‐1 trial. 1 Recombinant granulocyte colony‐stimulating factor (G‐CSF) has been widely used to shorten the duration of neutropenia and risk of infection in other settings, such as neutropenic complications after conditioning for hematopoietic cell transplant. 2 However, in CAR T‐cell therapy, myeloid growth factors have the potential to increase the incidence and/or severity of CRS and immune effector cell‐associated neurotoxicity syndrome (ICANS) by promotion of proinflammatory cytokine secretion from monocytes and macrophages. 3 , 4 , 5 Preclinically, monocytes and macrophages are the main source of interleukin (IL)‐1 and IL‐6 during CRS. 4 , 5 Surprisingly, CRS severity is mediated not by CAR T‐cell‐derived cytokines, but by those of monocytes and macrophages. 4 , 5 Therefore, we sought to determine if a relationship exists between filgrastim administration, which stimulates myelopoiesis and enhances granulocyte function, and CRS. There is currently a lack of evidence to guide clinicians on the benefits and risks of recombinant myeloid growth factor use in CAR T‐cell treatment. 2 MATERIALS AND METHODS Between March 2018 and May 2019, we reviewed 22 patients with DLBCL treated with axicabtagene ciloleucel with or without concurrent use of filgrastim. Prior to CAR T‐cell infusion, all patients received standard lymphodepleting therapy with fludarabine 30 mg/m2/day and cyclophosphamide 500 mg/m2/day on days −5 through −3, except for one patient that received reduced doses for chronic kidney disease Stage IV (fludarabine 15 mg/m2/day and cyclophosphamide 375 mg/m2/day). Prophylactic tocilizumab 8 mg/kg was given to all patients at 36 hours after CAR T‐cell infusion, with additional doses of tocilizumab and/or steroids given for evidence of CRS/ICANS based on the American Society for Transplantation and Cellular Therapy (ASTCT) consensus grading system. 6 Filgrastim was administered at physician discretion after CAR T‐cell infusion at a weight‐based dose of either 300 or 480 mcg, and cumulative filgrastim dose was recorded within the first 30 days. Toxicity after CAR T‐cell therapy was also assessed up to 30 days postinfusion. Any patient with neutropenic fever was treated with broad‐spectrum antibiotics; no prophylactic antibiotics were given. Documented infections were graded according to Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. 7 Antibiotics were also administered for any confirmed infections. Chi‐squared test or Cochran‐Armitage test were performed to examine association between cohort characteristics and the study variable of administration with or without filgrastim. 3 RESULTS Baseline patient characteristics are displayed in Table 1. The median age at the time of CAR T‐cell therapy was 65.0 years (interquartile range [IQR], 57.0‐68.8). The majority (14 patients, 63.6%) had high‐grade DLBCL, while 5 patients had DLBCL not otherwise specified, 2 patients had transformed follicular lymphoma and 1 patient had Richter's transformation. Seven patients (31.8%) had refractory DLBCL, while 15 patients (68.2%) had relapsed DLBCL with a median number of relapses prior to CAR T‐cell therapy of 1.0 (IQR, 0.0‐2.0). Thirteen patients (59.1%) had received greater than or equal to three prior therapies. Only one patient (4.6%) had received autologous hematopoietic cell transplant prior to CAR T‐cell therapy. Ten patients (45.5%) received bridging therapy for high disease burden before CAR T‐cell therapy, which included rituximab/gemcitabine‐oxaliplatin (7 patients), cytarabine/thiotepa (1 patient), rituximab/dexamethasone/cytarabine/cisplatin (1 patient), rituximab/ifosfamide/carboplatin/etoposide (RICE, 1 patient) and rituximab/cyclophosphamide/dexamethasone (1 patient). The number of cycles of bridging therapy ranged from 1 to 3 with a median of 2. TABLE 1 Baseline patient and disease characteristics All patients (N = 22) Age at time of CAR T‐cell therapy (median, IQR) 65.0 (57.0, 68.8) ≥65 12 (54.6%) <65 10 (45.5%) Gender Male 12 (54.6%) Female 10 (45.5%) Ethnicity Caucasian 12 (54.6%) Hispanic 7 (31.8%) Asian 0 (0%) African American 1 (4.6%) Other 2 (9.1%) Disease type DLBCL NOS 5 (22.7%) Transformed follicular 2 (9.1%) Richter's transformation 1 (4.6%) High‐grade DLBCL 14 (63.6%) Cell of origin GCB 10 (45.5%) Non‐GCB 10 (45.5%) Unspecified 2 (9.1%) Relapse or Refractory Relapse 15 (68.2%) Refractory 7 (31.8%) Number of prior therapies <3 9 (40.9%) ≥3 13 (59.1%) ASCT prior to CAR T‐cell therapy 1 (4.6%) ECOG at time of CAR T‐cell therapy 0‐1 21 (95.5%) 2‐4 1 (4.6%) Bridging therapy given before CAR T‐cell therapy Yes 10 (45.5%) No 12 (54.6%) Abbreviations: ASCT, autologous stem cell transplant; CAR, chimeric antigen receptor; DLBCL, diffuse large B‐cell lymphoma; ECOG, Eastern Cooperative Oncology Group; GCB, germinal center B‐cell; NOS, not otherwise specified. Seven of the 22 patients (31.8%) received filgrastim by physician discretion at a target dose of 5 mcg/kg/day with 3 patients receiving 300 mcg/day and 4 patients receiving 480 mcg/day. The median start day of filgrastim was 2 days post‐CAR T‐cell therapy (range Day −2 to Day +7), and the median number of filgrastim doses was 6 (range, 3‐18). The median duration of neutropenia after CAR T‐cell therapy was 5 days (IQR, 4.5‐8.5) for patients who received filgrastim compared to 15 days (IQR, 8.0‐30.0) for patients who did not receive filgrastim (P = .016; Table 2). Fourteen patients (63.6%) developed neutropenic fever after CAR T‐cell therapy, 6 of whom received filgrastim and 8 of whom had not (P = .193; Table 2). Five patients (22.7%) developed an infection in the 30 days post‐CAR T‐cell therapy including Clostridium difficile colitis (one patient, Grade 3), Enterococcus faecalis bacteremia (one patient, Grade 4), pneumonia (one patient, Grade 3) and Herpes simplex virus (two patients, Grades 1 and 2). Among the seven patients that received filgrastim, three patients (42.9%) developed an infection and two patients (28.6%) developed a Grade 3 or greater infection. Among the 15 patients that did not receive filgrastim, 2 patients (13.2%) developed an infection and 1 patient (6.7%) developed a Grade 3 or greater infection. There was no difference in the incidence and severity of infection between patients who received filgrastim and those that did not (P = .274, P = .138; Table 2; Figure 1). TABLE 2 CAR T‐cell associated toxicity All patients (N = 22) G‐CSF administered (N = 7) G‐CSF not administered (N = 15) P value Median duration of neutropenia (days) (IQR) 10 (6.0, 25.8) 5 (4.5, 8.5) 15 (8.0, 30.0) .016 Febrile neutropenia No 8 (36.4%) 1 (14.3%) 7 (46.7%) .193 Yes 14 (63.6%) 6 (85.7%) 8 (53.5%) New infection .274 None 17 (77.3%) 4 (57.1%) 13 (86.7%) Any grade 5 (22.7%) 3 (42.9%) 2 (13.2%) CRS .193 None 8 (36.4%) 1 (14.3%) 7 (46.7%) Any grade 14 (63.6%) 6 (85.7%) 8 (53.3%) ICANS .647 None 11 (50.0%) 3 (42.9%) 8 (53.3%) Any grade 11 (50.0%) 4 (57.1%) 7 (46.7%) Steroids given .648 No 9 (40.9%) 2 (28.6%) 7 (46.7%) Yes 13 (59.1%) 5 (71.4%) 8 (53.3%) More than one dose of tocilizumab given .074 No 10 (45.5%) 1 (14.3%) 9 (60.0%) Yes 12 (54.6%) 6 (85.7%) 6 (40.0%) Abbreviations: CAR, chimeric antigen receptor; CRS, cytokine release syndrome; G‐CSF, granulocyte colony‐stimulating factor; ICANS, immune effector cell‐associated neurotoxicity syndrome. FIGURE 1 Severity of chimeric antigen receptor (CAR) T‐cell associated toxicities based on granulocyte colony‐stimulating factor (G‐CSF) use. ICANS, immune effector cell‐associated neurotoxicity syndrome [Color figure can be viewed at wileyonlinelibrary.com] CRS was noted in 14 patients overall (63.6%), and 4 patients (18.2%) had Grade 3 or higher CRS. Among the seven patients that received filgrastim, six patients (85.7%) had evidence of CRS and three patients (42.9%) had Grade 3 or higher CRS. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) had evidence of CRS and 1 patient (6.7%) had evidence of Grade 3 or higher CRS. ICANS was noted in 11 patients overall (50.0%), and 9 patients (40.9%) had Grade 3 or higher ICANS. Among the seven patients that received filgrastim, four patients (57.1%) had evidence of ICANS and three patients (42.9%) had evidence of Grade 3 or higher ICANS. Among the 15 patients that did not receive filgrastim, 7 patients (46.7%) had evidence of ICANS and 6 patients (40.0%) had Grade 3 or higher ICANS. There was no significant difference in the incidence of developing CRS (any grade) or ICANS (any grade) between the group of patients that received filgrastim and those that did not (P = .193, P = .647; Table 2). There was, however, a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042), but no increase in the severity of ICANS based on filgrastim use (P = .660; Figure 1). Thirteen patients (59.1%) received corticosteroids after CAR T‐cell treatment at a median cumulative dosage of 666.7 mg prednisone equivalents (IQR, 445.0‐933.3), with the majority (9 patients, 69.2%) receiving steroids in the first 5 days post‐CAR T‐cell infusion. Half of the patient cohort (12 patients, 54.6%) required at least one dose of tocilizumab in addition to the scheduled prophylactic dose. Among the seven patients that received filgrastim, 5 patients (71.4%) received corticosteroids and 6 patients (85.7%) received more than one dose of tocilizumab. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) received corticosteroids and 6 patients (40.0%) received more than one dose of tocilizumab. There was no association between filgrastim use and steroid use or administration of additional doses of tocilizumab (P = .648, P = .074). 4 DISCUSSION At our institution, filgrastim was administered after CAR T‐cell therapy at physician discretion with less than half of our patient cohort receiving filgrastim (seven patients, 31.8%). Importantly, although the median duration of neutropenia was significantly lower for patients who received filgrastim, there was no difference in the development or severity of infections (P = .274, P = .138). Instead, while patients given filgrastim were not more likely to develop CRS (P = .193), the severity of CRS was higher in those that received filgrastim (P = .042), as has been suggested by preclinical data modeling myeloid growth factors in combination with CAR T‐cells. 4 , 5 The development of CRS is related to activation of in vivo T‐cell expansion and the production of T‐cell effector cytokines, including IL‐6, IL‐10 and interferon (IFN)‐γ, 8 which are also downstream products of myeloid cells stimulated by filgrastim. One preclinical study showed that higher levels of murine G‐CSF correlated strongly with CRS severity and survival. 5 Elevated systemic levels of IL‐6 in particular have been associated with severe CRS, and IL‐6 receptor blockade with the monoclonal antibody tocilizumab is an important agent used to reduce CRS toxicity. 9 , 10 Of all the cytokines analyzed in the ZUMA‐2 study, only peak levels of granzyme B and granulocyte‐macrophage colony stimulating factor (GM‐CSF, a closely related protein to G‐CSF) were associated with severe CRS and severe ICANS. 11 CAR T‐cell associated neurotoxicity has been associated with elevations in similar inflammatory markers to CRS (IL‐6, IL‐10, IFNγ), in addition to higher serum levels of G‐CSF and GM‐CSF. 12 , 13 Mouse models of CRS and ICANS have suggested that the main driver of CAR T‐cell neurotoxicity is actually IL‐1 secretion from activated macrophages. 4 , 5 Unlike its efficacy in CRS, tocilizumab did not protect mice from lethal neurotoxicity, a finding analogous in humans; however the IL‐1 receptor antagonist anakinra abolished both CRS and neurotoxicity. 4 , 12 Interestingly, in addition to its stimulatory effects on granulocytes, filgrastim has immunomodulatory effects on other immune cells and can inhibit inflammatory cytokine production within monocytes or macrophages. 14 This may account for why we found no association with filgrastim and the development or severity of neurotoxicity (P = .647, P = .660). GM‐CSF, which induces both granulocytes and macrophages, may be more closely tied to neurotoxic effects and, in fact, GM‐CSF inhibition has been shown to reduce neuroinflammation and prevent CRS. 15 The guidelines on use of filgrastim and other growth factors with CAR T‐cell therapy are not standardized. While some recommend administration of myeloid growth factors once absolute neutrophil count (ANC) decreases to <500/μL and to continue until ANC increases to ≥1500/μL, 16 others recommend administration of filgrastim only if patients develop Grade 1 CRS at the same time as neutropenia or in the setting of neutropenic fever. 17 The use of myeloid growth factors across institutions employing CAR T‐cell therapy is also mixed. In an electronic survey on the current administrative, logistic and toxicity management practices of CAR T‐cell therapy across the United States by the ASTCT, out of 28 respondents, 46% used growth factor if allowed by product labeling, 29% never administered growth factor, 14% determined the use of growth factor on a patient‐specific basis and 11% administered growth factor to all patients. 18 Although our study is limited by its small sample size and retrospective nature, we suggest that myeloid growth factor administration be used with caution in patients undergoing CAR T‐cell therapy. We found an association between filgrastim use and CRS severity, suggesting that increasing the number of patients in the study would strengthen the association. However, the effect size of the association was small. Further studies are required to determine the safety of myeloid growth factors after CAR T‐cell therapy. CONFLICT OF INTEREST The authors declared no potential conflicts of interest. ETHICS STATEMENT This study was approved by the University of California Los Angeles Institutional Review Board. Informed consent was not required for this study. ACKNOWLEDGMENT This work was supported by National Institutes of Health Grant Number 1K08CA245483‐01. Myung Shin Sim was supported by NIH National Center for Advancing Translational Sciences (Grant 5UL1TR001881‐05). DATA AVAILABILITY STATEMENT Data are available on request from the authors.
AXICABTAGENE CILOLEUCEL, CYCLOPHOSPHAMIDE, FILGRASTIM, FLUDARABINE PHOSPHATE, TOCILIZUMAB
DrugsGivenReaction
CC BY-NC
33091961
18,449,659
2021-03-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Febrile neutropenia'.
Filgrastim associations with CAR T-cell therapy. Little is known about the benefits and risks of myeloid growth factor administration after chimeric antigen receptor (CAR) T-cell therapy for diffuse large B-cell lymphoma (DLBCL). We present a retrospective analysis among 22 relapsed/refractory DLBCL patients who received CAR T-cell therapy with axicabtagene ciloleucel. Filgrastim was administered by physician discretion to seven patients (31.8%), and the median duration of neutropenia after lymphodepleting therapy was significantly shorter for those patients who received filgrastim (5 vs 15 days, P = .016). Five patients (22.7%) developed infection in the 30 days post-CAR T-cell therapy with three patients being Grade 3 or higher. There was no difference in the incidence and severity of infection based on filgrastim use (P = .274, P = .138). Among the seven patients that received filgrastim, six patients (85.7%) and four patients (57.1%) had evidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), respectively. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) and 7 patients (46.7%) had evidence of CRS and ICANS, respectively. There was no significant difference in the incidence of developing CRS or ICANS between the group of patients that received filgrastim and those that did not (P = .193, P = .647). However, there was a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042). Filgrastim administration after CAR T-cell therapy may lead to an increase in severity of CRS without decreasing infection rates. Abbreviations ANCabsolute neutrophil count ASTCTAmerican Society of Transplantation and Cellular Therapy CARchimeric antigen receptor CTCAECommon Terminology Criteria for Adverse Events DLBCLdiffuse large B‐cell lymphoma G‐CSFgranulocyte colony‐stimulating factor GM‐CSFgranulocyte‐macrophage colony‐stimulating factor ICANSimmune effector cell‐associated neurotoxicity syndrome IFNinterferon ILinterleukin IQRinterquartile range RICErituximab/ifosfamide/carboplatin/etoposide 1 INTRODUCTION While chimeric antigen receptor (CAR) T‐cell therapy is an exciting advancement in the treatment of patients with relapsed/refractory diffuse large B‐cell lymphoma (DLBCL), its application is limited by associated toxicities, including cytokine release syndrome (CRS), neurotoxicity and severe cytopenias, with 78% of patients developing Grade 3 or higher neutropenia in the ZUMA‐1 trial. 1 Recombinant granulocyte colony‐stimulating factor (G‐CSF) has been widely used to shorten the duration of neutropenia and risk of infection in other settings, such as neutropenic complications after conditioning for hematopoietic cell transplant. 2 However, in CAR T‐cell therapy, myeloid growth factors have the potential to increase the incidence and/or severity of CRS and immune effector cell‐associated neurotoxicity syndrome (ICANS) by promotion of proinflammatory cytokine secretion from monocytes and macrophages. 3 , 4 , 5 Preclinically, monocytes and macrophages are the main source of interleukin (IL)‐1 and IL‐6 during CRS. 4 , 5 Surprisingly, CRS severity is mediated not by CAR T‐cell‐derived cytokines, but by those of monocytes and macrophages. 4 , 5 Therefore, we sought to determine if a relationship exists between filgrastim administration, which stimulates myelopoiesis and enhances granulocyte function, and CRS. There is currently a lack of evidence to guide clinicians on the benefits and risks of recombinant myeloid growth factor use in CAR T‐cell treatment. 2 MATERIALS AND METHODS Between March 2018 and May 2019, we reviewed 22 patients with DLBCL treated with axicabtagene ciloleucel with or without concurrent use of filgrastim. Prior to CAR T‐cell infusion, all patients received standard lymphodepleting therapy with fludarabine 30 mg/m2/day and cyclophosphamide 500 mg/m2/day on days −5 through −3, except for one patient that received reduced doses for chronic kidney disease Stage IV (fludarabine 15 mg/m2/day and cyclophosphamide 375 mg/m2/day). Prophylactic tocilizumab 8 mg/kg was given to all patients at 36 hours after CAR T‐cell infusion, with additional doses of tocilizumab and/or steroids given for evidence of CRS/ICANS based on the American Society for Transplantation and Cellular Therapy (ASTCT) consensus grading system. 6 Filgrastim was administered at physician discretion after CAR T‐cell infusion at a weight‐based dose of either 300 or 480 mcg, and cumulative filgrastim dose was recorded within the first 30 days. Toxicity after CAR T‐cell therapy was also assessed up to 30 days postinfusion. Any patient with neutropenic fever was treated with broad‐spectrum antibiotics; no prophylactic antibiotics were given. Documented infections were graded according to Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. 7 Antibiotics were also administered for any confirmed infections. Chi‐squared test or Cochran‐Armitage test were performed to examine association between cohort characteristics and the study variable of administration with or without filgrastim. 3 RESULTS Baseline patient characteristics are displayed in Table 1. The median age at the time of CAR T‐cell therapy was 65.0 years (interquartile range [IQR], 57.0‐68.8). The majority (14 patients, 63.6%) had high‐grade DLBCL, while 5 patients had DLBCL not otherwise specified, 2 patients had transformed follicular lymphoma and 1 patient had Richter's transformation. Seven patients (31.8%) had refractory DLBCL, while 15 patients (68.2%) had relapsed DLBCL with a median number of relapses prior to CAR T‐cell therapy of 1.0 (IQR, 0.0‐2.0). Thirteen patients (59.1%) had received greater than or equal to three prior therapies. Only one patient (4.6%) had received autologous hematopoietic cell transplant prior to CAR T‐cell therapy. Ten patients (45.5%) received bridging therapy for high disease burden before CAR T‐cell therapy, which included rituximab/gemcitabine‐oxaliplatin (7 patients), cytarabine/thiotepa (1 patient), rituximab/dexamethasone/cytarabine/cisplatin (1 patient), rituximab/ifosfamide/carboplatin/etoposide (RICE, 1 patient) and rituximab/cyclophosphamide/dexamethasone (1 patient). The number of cycles of bridging therapy ranged from 1 to 3 with a median of 2. TABLE 1 Baseline patient and disease characteristics All patients (N = 22) Age at time of CAR T‐cell therapy (median, IQR) 65.0 (57.0, 68.8) ≥65 12 (54.6%) <65 10 (45.5%) Gender Male 12 (54.6%) Female 10 (45.5%) Ethnicity Caucasian 12 (54.6%) Hispanic 7 (31.8%) Asian 0 (0%) African American 1 (4.6%) Other 2 (9.1%) Disease type DLBCL NOS 5 (22.7%) Transformed follicular 2 (9.1%) Richter's transformation 1 (4.6%) High‐grade DLBCL 14 (63.6%) Cell of origin GCB 10 (45.5%) Non‐GCB 10 (45.5%) Unspecified 2 (9.1%) Relapse or Refractory Relapse 15 (68.2%) Refractory 7 (31.8%) Number of prior therapies <3 9 (40.9%) ≥3 13 (59.1%) ASCT prior to CAR T‐cell therapy 1 (4.6%) ECOG at time of CAR T‐cell therapy 0‐1 21 (95.5%) 2‐4 1 (4.6%) Bridging therapy given before CAR T‐cell therapy Yes 10 (45.5%) No 12 (54.6%) Abbreviations: ASCT, autologous stem cell transplant; CAR, chimeric antigen receptor; DLBCL, diffuse large B‐cell lymphoma; ECOG, Eastern Cooperative Oncology Group; GCB, germinal center B‐cell; NOS, not otherwise specified. Seven of the 22 patients (31.8%) received filgrastim by physician discretion at a target dose of 5 mcg/kg/day with 3 patients receiving 300 mcg/day and 4 patients receiving 480 mcg/day. The median start day of filgrastim was 2 days post‐CAR T‐cell therapy (range Day −2 to Day +7), and the median number of filgrastim doses was 6 (range, 3‐18). The median duration of neutropenia after CAR T‐cell therapy was 5 days (IQR, 4.5‐8.5) for patients who received filgrastim compared to 15 days (IQR, 8.0‐30.0) for patients who did not receive filgrastim (P = .016; Table 2). Fourteen patients (63.6%) developed neutropenic fever after CAR T‐cell therapy, 6 of whom received filgrastim and 8 of whom had not (P = .193; Table 2). Five patients (22.7%) developed an infection in the 30 days post‐CAR T‐cell therapy including Clostridium difficile colitis (one patient, Grade 3), Enterococcus faecalis bacteremia (one patient, Grade 4), pneumonia (one patient, Grade 3) and Herpes simplex virus (two patients, Grades 1 and 2). Among the seven patients that received filgrastim, three patients (42.9%) developed an infection and two patients (28.6%) developed a Grade 3 or greater infection. Among the 15 patients that did not receive filgrastim, 2 patients (13.2%) developed an infection and 1 patient (6.7%) developed a Grade 3 or greater infection. There was no difference in the incidence and severity of infection between patients who received filgrastim and those that did not (P = .274, P = .138; Table 2; Figure 1). TABLE 2 CAR T‐cell associated toxicity All patients (N = 22) G‐CSF administered (N = 7) G‐CSF not administered (N = 15) P value Median duration of neutropenia (days) (IQR) 10 (6.0, 25.8) 5 (4.5, 8.5) 15 (8.0, 30.0) .016 Febrile neutropenia No 8 (36.4%) 1 (14.3%) 7 (46.7%) .193 Yes 14 (63.6%) 6 (85.7%) 8 (53.5%) New infection .274 None 17 (77.3%) 4 (57.1%) 13 (86.7%) Any grade 5 (22.7%) 3 (42.9%) 2 (13.2%) CRS .193 None 8 (36.4%) 1 (14.3%) 7 (46.7%) Any grade 14 (63.6%) 6 (85.7%) 8 (53.3%) ICANS .647 None 11 (50.0%) 3 (42.9%) 8 (53.3%) Any grade 11 (50.0%) 4 (57.1%) 7 (46.7%) Steroids given .648 No 9 (40.9%) 2 (28.6%) 7 (46.7%) Yes 13 (59.1%) 5 (71.4%) 8 (53.3%) More than one dose of tocilizumab given .074 No 10 (45.5%) 1 (14.3%) 9 (60.0%) Yes 12 (54.6%) 6 (85.7%) 6 (40.0%) Abbreviations: CAR, chimeric antigen receptor; CRS, cytokine release syndrome; G‐CSF, granulocyte colony‐stimulating factor; ICANS, immune effector cell‐associated neurotoxicity syndrome. FIGURE 1 Severity of chimeric antigen receptor (CAR) T‐cell associated toxicities based on granulocyte colony‐stimulating factor (G‐CSF) use. ICANS, immune effector cell‐associated neurotoxicity syndrome [Color figure can be viewed at wileyonlinelibrary.com] CRS was noted in 14 patients overall (63.6%), and 4 patients (18.2%) had Grade 3 or higher CRS. Among the seven patients that received filgrastim, six patients (85.7%) had evidence of CRS and three patients (42.9%) had Grade 3 or higher CRS. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) had evidence of CRS and 1 patient (6.7%) had evidence of Grade 3 or higher CRS. ICANS was noted in 11 patients overall (50.0%), and 9 patients (40.9%) had Grade 3 or higher ICANS. Among the seven patients that received filgrastim, four patients (57.1%) had evidence of ICANS and three patients (42.9%) had evidence of Grade 3 or higher ICANS. Among the 15 patients that did not receive filgrastim, 7 patients (46.7%) had evidence of ICANS and 6 patients (40.0%) had Grade 3 or higher ICANS. There was no significant difference in the incidence of developing CRS (any grade) or ICANS (any grade) between the group of patients that received filgrastim and those that did not (P = .193, P = .647; Table 2). There was, however, a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042), but no increase in the severity of ICANS based on filgrastim use (P = .660; Figure 1). Thirteen patients (59.1%) received corticosteroids after CAR T‐cell treatment at a median cumulative dosage of 666.7 mg prednisone equivalents (IQR, 445.0‐933.3), with the majority (9 patients, 69.2%) receiving steroids in the first 5 days post‐CAR T‐cell infusion. Half of the patient cohort (12 patients, 54.6%) required at least one dose of tocilizumab in addition to the scheduled prophylactic dose. Among the seven patients that received filgrastim, 5 patients (71.4%) received corticosteroids and 6 patients (85.7%) received more than one dose of tocilizumab. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) received corticosteroids and 6 patients (40.0%) received more than one dose of tocilizumab. There was no association between filgrastim use and steroid use or administration of additional doses of tocilizumab (P = .648, P = .074). 4 DISCUSSION At our institution, filgrastim was administered after CAR T‐cell therapy at physician discretion with less than half of our patient cohort receiving filgrastim (seven patients, 31.8%). Importantly, although the median duration of neutropenia was significantly lower for patients who received filgrastim, there was no difference in the development or severity of infections (P = .274, P = .138). Instead, while patients given filgrastim were not more likely to develop CRS (P = .193), the severity of CRS was higher in those that received filgrastim (P = .042), as has been suggested by preclinical data modeling myeloid growth factors in combination with CAR T‐cells. 4 , 5 The development of CRS is related to activation of in vivo T‐cell expansion and the production of T‐cell effector cytokines, including IL‐6, IL‐10 and interferon (IFN)‐γ, 8 which are also downstream products of myeloid cells stimulated by filgrastim. One preclinical study showed that higher levels of murine G‐CSF correlated strongly with CRS severity and survival. 5 Elevated systemic levels of IL‐6 in particular have been associated with severe CRS, and IL‐6 receptor blockade with the monoclonal antibody tocilizumab is an important agent used to reduce CRS toxicity. 9 , 10 Of all the cytokines analyzed in the ZUMA‐2 study, only peak levels of granzyme B and granulocyte‐macrophage colony stimulating factor (GM‐CSF, a closely related protein to G‐CSF) were associated with severe CRS and severe ICANS. 11 CAR T‐cell associated neurotoxicity has been associated with elevations in similar inflammatory markers to CRS (IL‐6, IL‐10, IFNγ), in addition to higher serum levels of G‐CSF and GM‐CSF. 12 , 13 Mouse models of CRS and ICANS have suggested that the main driver of CAR T‐cell neurotoxicity is actually IL‐1 secretion from activated macrophages. 4 , 5 Unlike its efficacy in CRS, tocilizumab did not protect mice from lethal neurotoxicity, a finding analogous in humans; however the IL‐1 receptor antagonist anakinra abolished both CRS and neurotoxicity. 4 , 12 Interestingly, in addition to its stimulatory effects on granulocytes, filgrastim has immunomodulatory effects on other immune cells and can inhibit inflammatory cytokine production within monocytes or macrophages. 14 This may account for why we found no association with filgrastim and the development or severity of neurotoxicity (P = .647, P = .660). GM‐CSF, which induces both granulocytes and macrophages, may be more closely tied to neurotoxic effects and, in fact, GM‐CSF inhibition has been shown to reduce neuroinflammation and prevent CRS. 15 The guidelines on use of filgrastim and other growth factors with CAR T‐cell therapy are not standardized. While some recommend administration of myeloid growth factors once absolute neutrophil count (ANC) decreases to <500/μL and to continue until ANC increases to ≥1500/μL, 16 others recommend administration of filgrastim only if patients develop Grade 1 CRS at the same time as neutropenia or in the setting of neutropenic fever. 17 The use of myeloid growth factors across institutions employing CAR T‐cell therapy is also mixed. In an electronic survey on the current administrative, logistic and toxicity management practices of CAR T‐cell therapy across the United States by the ASTCT, out of 28 respondents, 46% used growth factor if allowed by product labeling, 29% never administered growth factor, 14% determined the use of growth factor on a patient‐specific basis and 11% administered growth factor to all patients. 18 Although our study is limited by its small sample size and retrospective nature, we suggest that myeloid growth factor administration be used with caution in patients undergoing CAR T‐cell therapy. We found an association between filgrastim use and CRS severity, suggesting that increasing the number of patients in the study would strengthen the association. However, the effect size of the association was small. Further studies are required to determine the safety of myeloid growth factors after CAR T‐cell therapy. CONFLICT OF INTEREST The authors declared no potential conflicts of interest. ETHICS STATEMENT This study was approved by the University of California Los Angeles Institutional Review Board. Informed consent was not required for this study. ACKNOWLEDGMENT This work was supported by National Institutes of Health Grant Number 1K08CA245483‐01. Myung Shin Sim was supported by NIH National Center for Advancing Translational Sciences (Grant 5UL1TR001881‐05). DATA AVAILABILITY STATEMENT Data are available on request from the authors.
AXICABTAGENE CILOLEUCEL, CYCLOPHOSPHAMIDE, FILGRASTIM, FLUDARABINE PHOSPHATE, TOCILIZUMAB
DrugsGivenReaction
CC BY-NC
33091961
18,449,659
2021-03-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Immune effector cell-associated neurotoxicity syndrome'.
Filgrastim associations with CAR T-cell therapy. Little is known about the benefits and risks of myeloid growth factor administration after chimeric antigen receptor (CAR) T-cell therapy for diffuse large B-cell lymphoma (DLBCL). We present a retrospective analysis among 22 relapsed/refractory DLBCL patients who received CAR T-cell therapy with axicabtagene ciloleucel. Filgrastim was administered by physician discretion to seven patients (31.8%), and the median duration of neutropenia after lymphodepleting therapy was significantly shorter for those patients who received filgrastim (5 vs 15 days, P = .016). Five patients (22.7%) developed infection in the 30 days post-CAR T-cell therapy with three patients being Grade 3 or higher. There was no difference in the incidence and severity of infection based on filgrastim use (P = .274, P = .138). Among the seven patients that received filgrastim, six patients (85.7%) and four patients (57.1%) had evidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), respectively. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) and 7 patients (46.7%) had evidence of CRS and ICANS, respectively. There was no significant difference in the incidence of developing CRS or ICANS between the group of patients that received filgrastim and those that did not (P = .193, P = .647). However, there was a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042). Filgrastim administration after CAR T-cell therapy may lead to an increase in severity of CRS without decreasing infection rates. Abbreviations ANCabsolute neutrophil count ASTCTAmerican Society of Transplantation and Cellular Therapy CARchimeric antigen receptor CTCAECommon Terminology Criteria for Adverse Events DLBCLdiffuse large B‐cell lymphoma G‐CSFgranulocyte colony‐stimulating factor GM‐CSFgranulocyte‐macrophage colony‐stimulating factor ICANSimmune effector cell‐associated neurotoxicity syndrome IFNinterferon ILinterleukin IQRinterquartile range RICErituximab/ifosfamide/carboplatin/etoposide 1 INTRODUCTION While chimeric antigen receptor (CAR) T‐cell therapy is an exciting advancement in the treatment of patients with relapsed/refractory diffuse large B‐cell lymphoma (DLBCL), its application is limited by associated toxicities, including cytokine release syndrome (CRS), neurotoxicity and severe cytopenias, with 78% of patients developing Grade 3 or higher neutropenia in the ZUMA‐1 trial. 1 Recombinant granulocyte colony‐stimulating factor (G‐CSF) has been widely used to shorten the duration of neutropenia and risk of infection in other settings, such as neutropenic complications after conditioning for hematopoietic cell transplant. 2 However, in CAR T‐cell therapy, myeloid growth factors have the potential to increase the incidence and/or severity of CRS and immune effector cell‐associated neurotoxicity syndrome (ICANS) by promotion of proinflammatory cytokine secretion from monocytes and macrophages. 3 , 4 , 5 Preclinically, monocytes and macrophages are the main source of interleukin (IL)‐1 and IL‐6 during CRS. 4 , 5 Surprisingly, CRS severity is mediated not by CAR T‐cell‐derived cytokines, but by those of monocytes and macrophages. 4 , 5 Therefore, we sought to determine if a relationship exists between filgrastim administration, which stimulates myelopoiesis and enhances granulocyte function, and CRS. There is currently a lack of evidence to guide clinicians on the benefits and risks of recombinant myeloid growth factor use in CAR T‐cell treatment. 2 MATERIALS AND METHODS Between March 2018 and May 2019, we reviewed 22 patients with DLBCL treated with axicabtagene ciloleucel with or without concurrent use of filgrastim. Prior to CAR T‐cell infusion, all patients received standard lymphodepleting therapy with fludarabine 30 mg/m2/day and cyclophosphamide 500 mg/m2/day on days −5 through −3, except for one patient that received reduced doses for chronic kidney disease Stage IV (fludarabine 15 mg/m2/day and cyclophosphamide 375 mg/m2/day). Prophylactic tocilizumab 8 mg/kg was given to all patients at 36 hours after CAR T‐cell infusion, with additional doses of tocilizumab and/or steroids given for evidence of CRS/ICANS based on the American Society for Transplantation and Cellular Therapy (ASTCT) consensus grading system. 6 Filgrastim was administered at physician discretion after CAR T‐cell infusion at a weight‐based dose of either 300 or 480 mcg, and cumulative filgrastim dose was recorded within the first 30 days. Toxicity after CAR T‐cell therapy was also assessed up to 30 days postinfusion. Any patient with neutropenic fever was treated with broad‐spectrum antibiotics; no prophylactic antibiotics were given. Documented infections were graded according to Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. 7 Antibiotics were also administered for any confirmed infections. Chi‐squared test or Cochran‐Armitage test were performed to examine association between cohort characteristics and the study variable of administration with or without filgrastim. 3 RESULTS Baseline patient characteristics are displayed in Table 1. The median age at the time of CAR T‐cell therapy was 65.0 years (interquartile range [IQR], 57.0‐68.8). The majority (14 patients, 63.6%) had high‐grade DLBCL, while 5 patients had DLBCL not otherwise specified, 2 patients had transformed follicular lymphoma and 1 patient had Richter's transformation. Seven patients (31.8%) had refractory DLBCL, while 15 patients (68.2%) had relapsed DLBCL with a median number of relapses prior to CAR T‐cell therapy of 1.0 (IQR, 0.0‐2.0). Thirteen patients (59.1%) had received greater than or equal to three prior therapies. Only one patient (4.6%) had received autologous hematopoietic cell transplant prior to CAR T‐cell therapy. Ten patients (45.5%) received bridging therapy for high disease burden before CAR T‐cell therapy, which included rituximab/gemcitabine‐oxaliplatin (7 patients), cytarabine/thiotepa (1 patient), rituximab/dexamethasone/cytarabine/cisplatin (1 patient), rituximab/ifosfamide/carboplatin/etoposide (RICE, 1 patient) and rituximab/cyclophosphamide/dexamethasone (1 patient). The number of cycles of bridging therapy ranged from 1 to 3 with a median of 2. TABLE 1 Baseline patient and disease characteristics All patients (N = 22) Age at time of CAR T‐cell therapy (median, IQR) 65.0 (57.0, 68.8) ≥65 12 (54.6%) <65 10 (45.5%) Gender Male 12 (54.6%) Female 10 (45.5%) Ethnicity Caucasian 12 (54.6%) Hispanic 7 (31.8%) Asian 0 (0%) African American 1 (4.6%) Other 2 (9.1%) Disease type DLBCL NOS 5 (22.7%) Transformed follicular 2 (9.1%) Richter's transformation 1 (4.6%) High‐grade DLBCL 14 (63.6%) Cell of origin GCB 10 (45.5%) Non‐GCB 10 (45.5%) Unspecified 2 (9.1%) Relapse or Refractory Relapse 15 (68.2%) Refractory 7 (31.8%) Number of prior therapies <3 9 (40.9%) ≥3 13 (59.1%) ASCT prior to CAR T‐cell therapy 1 (4.6%) ECOG at time of CAR T‐cell therapy 0‐1 21 (95.5%) 2‐4 1 (4.6%) Bridging therapy given before CAR T‐cell therapy Yes 10 (45.5%) No 12 (54.6%) Abbreviations: ASCT, autologous stem cell transplant; CAR, chimeric antigen receptor; DLBCL, diffuse large B‐cell lymphoma; ECOG, Eastern Cooperative Oncology Group; GCB, germinal center B‐cell; NOS, not otherwise specified. Seven of the 22 patients (31.8%) received filgrastim by physician discretion at a target dose of 5 mcg/kg/day with 3 patients receiving 300 mcg/day and 4 patients receiving 480 mcg/day. The median start day of filgrastim was 2 days post‐CAR T‐cell therapy (range Day −2 to Day +7), and the median number of filgrastim doses was 6 (range, 3‐18). The median duration of neutropenia after CAR T‐cell therapy was 5 days (IQR, 4.5‐8.5) for patients who received filgrastim compared to 15 days (IQR, 8.0‐30.0) for patients who did not receive filgrastim (P = .016; Table 2). Fourteen patients (63.6%) developed neutropenic fever after CAR T‐cell therapy, 6 of whom received filgrastim and 8 of whom had not (P = .193; Table 2). Five patients (22.7%) developed an infection in the 30 days post‐CAR T‐cell therapy including Clostridium difficile colitis (one patient, Grade 3), Enterococcus faecalis bacteremia (one patient, Grade 4), pneumonia (one patient, Grade 3) and Herpes simplex virus (two patients, Grades 1 and 2). Among the seven patients that received filgrastim, three patients (42.9%) developed an infection and two patients (28.6%) developed a Grade 3 or greater infection. Among the 15 patients that did not receive filgrastim, 2 patients (13.2%) developed an infection and 1 patient (6.7%) developed a Grade 3 or greater infection. There was no difference in the incidence and severity of infection between patients who received filgrastim and those that did not (P = .274, P = .138; Table 2; Figure 1). TABLE 2 CAR T‐cell associated toxicity All patients (N = 22) G‐CSF administered (N = 7) G‐CSF not administered (N = 15) P value Median duration of neutropenia (days) (IQR) 10 (6.0, 25.8) 5 (4.5, 8.5) 15 (8.0, 30.0) .016 Febrile neutropenia No 8 (36.4%) 1 (14.3%) 7 (46.7%) .193 Yes 14 (63.6%) 6 (85.7%) 8 (53.5%) New infection .274 None 17 (77.3%) 4 (57.1%) 13 (86.7%) Any grade 5 (22.7%) 3 (42.9%) 2 (13.2%) CRS .193 None 8 (36.4%) 1 (14.3%) 7 (46.7%) Any grade 14 (63.6%) 6 (85.7%) 8 (53.3%) ICANS .647 None 11 (50.0%) 3 (42.9%) 8 (53.3%) Any grade 11 (50.0%) 4 (57.1%) 7 (46.7%) Steroids given .648 No 9 (40.9%) 2 (28.6%) 7 (46.7%) Yes 13 (59.1%) 5 (71.4%) 8 (53.3%) More than one dose of tocilizumab given .074 No 10 (45.5%) 1 (14.3%) 9 (60.0%) Yes 12 (54.6%) 6 (85.7%) 6 (40.0%) Abbreviations: CAR, chimeric antigen receptor; CRS, cytokine release syndrome; G‐CSF, granulocyte colony‐stimulating factor; ICANS, immune effector cell‐associated neurotoxicity syndrome. FIGURE 1 Severity of chimeric antigen receptor (CAR) T‐cell associated toxicities based on granulocyte colony‐stimulating factor (G‐CSF) use. ICANS, immune effector cell‐associated neurotoxicity syndrome [Color figure can be viewed at wileyonlinelibrary.com] CRS was noted in 14 patients overall (63.6%), and 4 patients (18.2%) had Grade 3 or higher CRS. Among the seven patients that received filgrastim, six patients (85.7%) had evidence of CRS and three patients (42.9%) had Grade 3 or higher CRS. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) had evidence of CRS and 1 patient (6.7%) had evidence of Grade 3 or higher CRS. ICANS was noted in 11 patients overall (50.0%), and 9 patients (40.9%) had Grade 3 or higher ICANS. Among the seven patients that received filgrastim, four patients (57.1%) had evidence of ICANS and three patients (42.9%) had evidence of Grade 3 or higher ICANS. Among the 15 patients that did not receive filgrastim, 7 patients (46.7%) had evidence of ICANS and 6 patients (40.0%) had Grade 3 or higher ICANS. There was no significant difference in the incidence of developing CRS (any grade) or ICANS (any grade) between the group of patients that received filgrastim and those that did not (P = .193, P = .647; Table 2). There was, however, a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042), but no increase in the severity of ICANS based on filgrastim use (P = .660; Figure 1). Thirteen patients (59.1%) received corticosteroids after CAR T‐cell treatment at a median cumulative dosage of 666.7 mg prednisone equivalents (IQR, 445.0‐933.3), with the majority (9 patients, 69.2%) receiving steroids in the first 5 days post‐CAR T‐cell infusion. Half of the patient cohort (12 patients, 54.6%) required at least one dose of tocilizumab in addition to the scheduled prophylactic dose. Among the seven patients that received filgrastim, 5 patients (71.4%) received corticosteroids and 6 patients (85.7%) received more than one dose of tocilizumab. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) received corticosteroids and 6 patients (40.0%) received more than one dose of tocilizumab. There was no association between filgrastim use and steroid use or administration of additional doses of tocilizumab (P = .648, P = .074). 4 DISCUSSION At our institution, filgrastim was administered after CAR T‐cell therapy at physician discretion with less than half of our patient cohort receiving filgrastim (seven patients, 31.8%). Importantly, although the median duration of neutropenia was significantly lower for patients who received filgrastim, there was no difference in the development or severity of infections (P = .274, P = .138). Instead, while patients given filgrastim were not more likely to develop CRS (P = .193), the severity of CRS was higher in those that received filgrastim (P = .042), as has been suggested by preclinical data modeling myeloid growth factors in combination with CAR T‐cells. 4 , 5 The development of CRS is related to activation of in vivo T‐cell expansion and the production of T‐cell effector cytokines, including IL‐6, IL‐10 and interferon (IFN)‐γ, 8 which are also downstream products of myeloid cells stimulated by filgrastim. One preclinical study showed that higher levels of murine G‐CSF correlated strongly with CRS severity and survival. 5 Elevated systemic levels of IL‐6 in particular have been associated with severe CRS, and IL‐6 receptor blockade with the monoclonal antibody tocilizumab is an important agent used to reduce CRS toxicity. 9 , 10 Of all the cytokines analyzed in the ZUMA‐2 study, only peak levels of granzyme B and granulocyte‐macrophage colony stimulating factor (GM‐CSF, a closely related protein to G‐CSF) were associated with severe CRS and severe ICANS. 11 CAR T‐cell associated neurotoxicity has been associated with elevations in similar inflammatory markers to CRS (IL‐6, IL‐10, IFNγ), in addition to higher serum levels of G‐CSF and GM‐CSF. 12 , 13 Mouse models of CRS and ICANS have suggested that the main driver of CAR T‐cell neurotoxicity is actually IL‐1 secretion from activated macrophages. 4 , 5 Unlike its efficacy in CRS, tocilizumab did not protect mice from lethal neurotoxicity, a finding analogous in humans; however the IL‐1 receptor antagonist anakinra abolished both CRS and neurotoxicity. 4 , 12 Interestingly, in addition to its stimulatory effects on granulocytes, filgrastim has immunomodulatory effects on other immune cells and can inhibit inflammatory cytokine production within monocytes or macrophages. 14 This may account for why we found no association with filgrastim and the development or severity of neurotoxicity (P = .647, P = .660). GM‐CSF, which induces both granulocytes and macrophages, may be more closely tied to neurotoxic effects and, in fact, GM‐CSF inhibition has been shown to reduce neuroinflammation and prevent CRS. 15 The guidelines on use of filgrastim and other growth factors with CAR T‐cell therapy are not standardized. While some recommend administration of myeloid growth factors once absolute neutrophil count (ANC) decreases to <500/μL and to continue until ANC increases to ≥1500/μL, 16 others recommend administration of filgrastim only if patients develop Grade 1 CRS at the same time as neutropenia or in the setting of neutropenic fever. 17 The use of myeloid growth factors across institutions employing CAR T‐cell therapy is also mixed. In an electronic survey on the current administrative, logistic and toxicity management practices of CAR T‐cell therapy across the United States by the ASTCT, out of 28 respondents, 46% used growth factor if allowed by product labeling, 29% never administered growth factor, 14% determined the use of growth factor on a patient‐specific basis and 11% administered growth factor to all patients. 18 Although our study is limited by its small sample size and retrospective nature, we suggest that myeloid growth factor administration be used with caution in patients undergoing CAR T‐cell therapy. We found an association between filgrastim use and CRS severity, suggesting that increasing the number of patients in the study would strengthen the association. However, the effect size of the association was small. Further studies are required to determine the safety of myeloid growth factors after CAR T‐cell therapy. CONFLICT OF INTEREST The authors declared no potential conflicts of interest. ETHICS STATEMENT This study was approved by the University of California Los Angeles Institutional Review Board. Informed consent was not required for this study. ACKNOWLEDGMENT This work was supported by National Institutes of Health Grant Number 1K08CA245483‐01. Myung Shin Sim was supported by NIH National Center for Advancing Translational Sciences (Grant 5UL1TR001881‐05). DATA AVAILABILITY STATEMENT Data are available on request from the authors.
AXICABTAGENE CILOLEUCEL, CYCLOPHOSPHAMIDE, FILGRASTIM, FLUDARABINE PHOSPHATE, TOCILIZUMAB
DrugsGivenReaction
CC BY-NC
33091961
18,449,659
2021-03-01
Give an alphabetized list of all active substances of drugs taken by the patient who experienced 'Infection'.
Filgrastim associations with CAR T-cell therapy. Little is known about the benefits and risks of myeloid growth factor administration after chimeric antigen receptor (CAR) T-cell therapy for diffuse large B-cell lymphoma (DLBCL). We present a retrospective analysis among 22 relapsed/refractory DLBCL patients who received CAR T-cell therapy with axicabtagene ciloleucel. Filgrastim was administered by physician discretion to seven patients (31.8%), and the median duration of neutropenia after lymphodepleting therapy was significantly shorter for those patients who received filgrastim (5 vs 15 days, P = .016). Five patients (22.7%) developed infection in the 30 days post-CAR T-cell therapy with three patients being Grade 3 or higher. There was no difference in the incidence and severity of infection based on filgrastim use (P = .274, P = .138). Among the seven patients that received filgrastim, six patients (85.7%) and four patients (57.1%) had evidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), respectively. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) and 7 patients (46.7%) had evidence of CRS and ICANS, respectively. There was no significant difference in the incidence of developing CRS or ICANS between the group of patients that received filgrastim and those that did not (P = .193, P = .647). However, there was a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042). Filgrastim administration after CAR T-cell therapy may lead to an increase in severity of CRS without decreasing infection rates. Abbreviations ANCabsolute neutrophil count ASTCTAmerican Society of Transplantation and Cellular Therapy CARchimeric antigen receptor CTCAECommon Terminology Criteria for Adverse Events DLBCLdiffuse large B‐cell lymphoma G‐CSFgranulocyte colony‐stimulating factor GM‐CSFgranulocyte‐macrophage colony‐stimulating factor ICANSimmune effector cell‐associated neurotoxicity syndrome IFNinterferon ILinterleukin IQRinterquartile range RICErituximab/ifosfamide/carboplatin/etoposide 1 INTRODUCTION While chimeric antigen receptor (CAR) T‐cell therapy is an exciting advancement in the treatment of patients with relapsed/refractory diffuse large B‐cell lymphoma (DLBCL), its application is limited by associated toxicities, including cytokine release syndrome (CRS), neurotoxicity and severe cytopenias, with 78% of patients developing Grade 3 or higher neutropenia in the ZUMA‐1 trial. 1 Recombinant granulocyte colony‐stimulating factor (G‐CSF) has been widely used to shorten the duration of neutropenia and risk of infection in other settings, such as neutropenic complications after conditioning for hematopoietic cell transplant. 2 However, in CAR T‐cell therapy, myeloid growth factors have the potential to increase the incidence and/or severity of CRS and immune effector cell‐associated neurotoxicity syndrome (ICANS) by promotion of proinflammatory cytokine secretion from monocytes and macrophages. 3 , 4 , 5 Preclinically, monocytes and macrophages are the main source of interleukin (IL)‐1 and IL‐6 during CRS. 4 , 5 Surprisingly, CRS severity is mediated not by CAR T‐cell‐derived cytokines, but by those of monocytes and macrophages. 4 , 5 Therefore, we sought to determine if a relationship exists between filgrastim administration, which stimulates myelopoiesis and enhances granulocyte function, and CRS. There is currently a lack of evidence to guide clinicians on the benefits and risks of recombinant myeloid growth factor use in CAR T‐cell treatment. 2 MATERIALS AND METHODS Between March 2018 and May 2019, we reviewed 22 patients with DLBCL treated with axicabtagene ciloleucel with or without concurrent use of filgrastim. Prior to CAR T‐cell infusion, all patients received standard lymphodepleting therapy with fludarabine 30 mg/m2/day and cyclophosphamide 500 mg/m2/day on days −5 through −3, except for one patient that received reduced doses for chronic kidney disease Stage IV (fludarabine 15 mg/m2/day and cyclophosphamide 375 mg/m2/day). Prophylactic tocilizumab 8 mg/kg was given to all patients at 36 hours after CAR T‐cell infusion, with additional doses of tocilizumab and/or steroids given for evidence of CRS/ICANS based on the American Society for Transplantation and Cellular Therapy (ASTCT) consensus grading system. 6 Filgrastim was administered at physician discretion after CAR T‐cell infusion at a weight‐based dose of either 300 or 480 mcg, and cumulative filgrastim dose was recorded within the first 30 days. Toxicity after CAR T‐cell therapy was also assessed up to 30 days postinfusion. Any patient with neutropenic fever was treated with broad‐spectrum antibiotics; no prophylactic antibiotics were given. Documented infections were graded according to Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. 7 Antibiotics were also administered for any confirmed infections. Chi‐squared test or Cochran‐Armitage test were performed to examine association between cohort characteristics and the study variable of administration with or without filgrastim. 3 RESULTS Baseline patient characteristics are displayed in Table 1. The median age at the time of CAR T‐cell therapy was 65.0 years (interquartile range [IQR], 57.0‐68.8). The majority (14 patients, 63.6%) had high‐grade DLBCL, while 5 patients had DLBCL not otherwise specified, 2 patients had transformed follicular lymphoma and 1 patient had Richter's transformation. Seven patients (31.8%) had refractory DLBCL, while 15 patients (68.2%) had relapsed DLBCL with a median number of relapses prior to CAR T‐cell therapy of 1.0 (IQR, 0.0‐2.0). Thirteen patients (59.1%) had received greater than or equal to three prior therapies. Only one patient (4.6%) had received autologous hematopoietic cell transplant prior to CAR T‐cell therapy. Ten patients (45.5%) received bridging therapy for high disease burden before CAR T‐cell therapy, which included rituximab/gemcitabine‐oxaliplatin (7 patients), cytarabine/thiotepa (1 patient), rituximab/dexamethasone/cytarabine/cisplatin (1 patient), rituximab/ifosfamide/carboplatin/etoposide (RICE, 1 patient) and rituximab/cyclophosphamide/dexamethasone (1 patient). The number of cycles of bridging therapy ranged from 1 to 3 with a median of 2. TABLE 1 Baseline patient and disease characteristics All patients (N = 22) Age at time of CAR T‐cell therapy (median, IQR) 65.0 (57.0, 68.8) ≥65 12 (54.6%) <65 10 (45.5%) Gender Male 12 (54.6%) Female 10 (45.5%) Ethnicity Caucasian 12 (54.6%) Hispanic 7 (31.8%) Asian 0 (0%) African American 1 (4.6%) Other 2 (9.1%) Disease type DLBCL NOS 5 (22.7%) Transformed follicular 2 (9.1%) Richter's transformation 1 (4.6%) High‐grade DLBCL 14 (63.6%) Cell of origin GCB 10 (45.5%) Non‐GCB 10 (45.5%) Unspecified 2 (9.1%) Relapse or Refractory Relapse 15 (68.2%) Refractory 7 (31.8%) Number of prior therapies <3 9 (40.9%) ≥3 13 (59.1%) ASCT prior to CAR T‐cell therapy 1 (4.6%) ECOG at time of CAR T‐cell therapy 0‐1 21 (95.5%) 2‐4 1 (4.6%) Bridging therapy given before CAR T‐cell therapy Yes 10 (45.5%) No 12 (54.6%) Abbreviations: ASCT, autologous stem cell transplant; CAR, chimeric antigen receptor; DLBCL, diffuse large B‐cell lymphoma; ECOG, Eastern Cooperative Oncology Group; GCB, germinal center B‐cell; NOS, not otherwise specified. Seven of the 22 patients (31.8%) received filgrastim by physician discretion at a target dose of 5 mcg/kg/day with 3 patients receiving 300 mcg/day and 4 patients receiving 480 mcg/day. The median start day of filgrastim was 2 days post‐CAR T‐cell therapy (range Day −2 to Day +7), and the median number of filgrastim doses was 6 (range, 3‐18). The median duration of neutropenia after CAR T‐cell therapy was 5 days (IQR, 4.5‐8.5) for patients who received filgrastim compared to 15 days (IQR, 8.0‐30.0) for patients who did not receive filgrastim (P = .016; Table 2). Fourteen patients (63.6%) developed neutropenic fever after CAR T‐cell therapy, 6 of whom received filgrastim and 8 of whom had not (P = .193; Table 2). Five patients (22.7%) developed an infection in the 30 days post‐CAR T‐cell therapy including Clostridium difficile colitis (one patient, Grade 3), Enterococcus faecalis bacteremia (one patient, Grade 4), pneumonia (one patient, Grade 3) and Herpes simplex virus (two patients, Grades 1 and 2). Among the seven patients that received filgrastim, three patients (42.9%) developed an infection and two patients (28.6%) developed a Grade 3 or greater infection. Among the 15 patients that did not receive filgrastim, 2 patients (13.2%) developed an infection and 1 patient (6.7%) developed a Grade 3 or greater infection. There was no difference in the incidence and severity of infection between patients who received filgrastim and those that did not (P = .274, P = .138; Table 2; Figure 1). TABLE 2 CAR T‐cell associated toxicity All patients (N = 22) G‐CSF administered (N = 7) G‐CSF not administered (N = 15) P value Median duration of neutropenia (days) (IQR) 10 (6.0, 25.8) 5 (4.5, 8.5) 15 (8.0, 30.0) .016 Febrile neutropenia No 8 (36.4%) 1 (14.3%) 7 (46.7%) .193 Yes 14 (63.6%) 6 (85.7%) 8 (53.5%) New infection .274 None 17 (77.3%) 4 (57.1%) 13 (86.7%) Any grade 5 (22.7%) 3 (42.9%) 2 (13.2%) CRS .193 None 8 (36.4%) 1 (14.3%) 7 (46.7%) Any grade 14 (63.6%) 6 (85.7%) 8 (53.3%) ICANS .647 None 11 (50.0%) 3 (42.9%) 8 (53.3%) Any grade 11 (50.0%) 4 (57.1%) 7 (46.7%) Steroids given .648 No 9 (40.9%) 2 (28.6%) 7 (46.7%) Yes 13 (59.1%) 5 (71.4%) 8 (53.3%) More than one dose of tocilizumab given .074 No 10 (45.5%) 1 (14.3%) 9 (60.0%) Yes 12 (54.6%) 6 (85.7%) 6 (40.0%) Abbreviations: CAR, chimeric antigen receptor; CRS, cytokine release syndrome; G‐CSF, granulocyte colony‐stimulating factor; ICANS, immune effector cell‐associated neurotoxicity syndrome. FIGURE 1 Severity of chimeric antigen receptor (CAR) T‐cell associated toxicities based on granulocyte colony‐stimulating factor (G‐CSF) use. ICANS, immune effector cell‐associated neurotoxicity syndrome [Color figure can be viewed at wileyonlinelibrary.com] CRS was noted in 14 patients overall (63.6%), and 4 patients (18.2%) had Grade 3 or higher CRS. Among the seven patients that received filgrastim, six patients (85.7%) had evidence of CRS and three patients (42.9%) had Grade 3 or higher CRS. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) had evidence of CRS and 1 patient (6.7%) had evidence of Grade 3 or higher CRS. ICANS was noted in 11 patients overall (50.0%), and 9 patients (40.9%) had Grade 3 or higher ICANS. Among the seven patients that received filgrastim, four patients (57.1%) had evidence of ICANS and three patients (42.9%) had evidence of Grade 3 or higher ICANS. Among the 15 patients that did not receive filgrastim, 7 patients (46.7%) had evidence of ICANS and 6 patients (40.0%) had Grade 3 or higher ICANS. There was no significant difference in the incidence of developing CRS (any grade) or ICANS (any grade) between the group of patients that received filgrastim and those that did not (P = .193, P = .647; Table 2). There was, however, a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042), but no increase in the severity of ICANS based on filgrastim use (P = .660; Figure 1). Thirteen patients (59.1%) received corticosteroids after CAR T‐cell treatment at a median cumulative dosage of 666.7 mg prednisone equivalents (IQR, 445.0‐933.3), with the majority (9 patients, 69.2%) receiving steroids in the first 5 days post‐CAR T‐cell infusion. Half of the patient cohort (12 patients, 54.6%) required at least one dose of tocilizumab in addition to the scheduled prophylactic dose. Among the seven patients that received filgrastim, 5 patients (71.4%) received corticosteroids and 6 patients (85.7%) received more than one dose of tocilizumab. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) received corticosteroids and 6 patients (40.0%) received more than one dose of tocilizumab. There was no association between filgrastim use and steroid use or administration of additional doses of tocilizumab (P = .648, P = .074). 4 DISCUSSION At our institution, filgrastim was administered after CAR T‐cell therapy at physician discretion with less than half of our patient cohort receiving filgrastim (seven patients, 31.8%). Importantly, although the median duration of neutropenia was significantly lower for patients who received filgrastim, there was no difference in the development or severity of infections (P = .274, P = .138). Instead, while patients given filgrastim were not more likely to develop CRS (P = .193), the severity of CRS was higher in those that received filgrastim (P = .042), as has been suggested by preclinical data modeling myeloid growth factors in combination with CAR T‐cells. 4 , 5 The development of CRS is related to activation of in vivo T‐cell expansion and the production of T‐cell effector cytokines, including IL‐6, IL‐10 and interferon (IFN)‐γ, 8 which are also downstream products of myeloid cells stimulated by filgrastim. One preclinical study showed that higher levels of murine G‐CSF correlated strongly with CRS severity and survival. 5 Elevated systemic levels of IL‐6 in particular have been associated with severe CRS, and IL‐6 receptor blockade with the monoclonal antibody tocilizumab is an important agent used to reduce CRS toxicity. 9 , 10 Of all the cytokines analyzed in the ZUMA‐2 study, only peak levels of granzyme B and granulocyte‐macrophage colony stimulating factor (GM‐CSF, a closely related protein to G‐CSF) were associated with severe CRS and severe ICANS. 11 CAR T‐cell associated neurotoxicity has been associated with elevations in similar inflammatory markers to CRS (IL‐6, IL‐10, IFNγ), in addition to higher serum levels of G‐CSF and GM‐CSF. 12 , 13 Mouse models of CRS and ICANS have suggested that the main driver of CAR T‐cell neurotoxicity is actually IL‐1 secretion from activated macrophages. 4 , 5 Unlike its efficacy in CRS, tocilizumab did not protect mice from lethal neurotoxicity, a finding analogous in humans; however the IL‐1 receptor antagonist anakinra abolished both CRS and neurotoxicity. 4 , 12 Interestingly, in addition to its stimulatory effects on granulocytes, filgrastim has immunomodulatory effects on other immune cells and can inhibit inflammatory cytokine production within monocytes or macrophages. 14 This may account for why we found no association with filgrastim and the development or severity of neurotoxicity (P = .647, P = .660). GM‐CSF, which induces both granulocytes and macrophages, may be more closely tied to neurotoxic effects and, in fact, GM‐CSF inhibition has been shown to reduce neuroinflammation and prevent CRS. 15 The guidelines on use of filgrastim and other growth factors with CAR T‐cell therapy are not standardized. While some recommend administration of myeloid growth factors once absolute neutrophil count (ANC) decreases to <500/μL and to continue until ANC increases to ≥1500/μL, 16 others recommend administration of filgrastim only if patients develop Grade 1 CRS at the same time as neutropenia or in the setting of neutropenic fever. 17 The use of myeloid growth factors across institutions employing CAR T‐cell therapy is also mixed. In an electronic survey on the current administrative, logistic and toxicity management practices of CAR T‐cell therapy across the United States by the ASTCT, out of 28 respondents, 46% used growth factor if allowed by product labeling, 29% never administered growth factor, 14% determined the use of growth factor on a patient‐specific basis and 11% administered growth factor to all patients. 18 Although our study is limited by its small sample size and retrospective nature, we suggest that myeloid growth factor administration be used with caution in patients undergoing CAR T‐cell therapy. We found an association between filgrastim use and CRS severity, suggesting that increasing the number of patients in the study would strengthen the association. However, the effect size of the association was small. Further studies are required to determine the safety of myeloid growth factors after CAR T‐cell therapy. CONFLICT OF INTEREST The authors declared no potential conflicts of interest. ETHICS STATEMENT This study was approved by the University of California Los Angeles Institutional Review Board. Informed consent was not required for this study. ACKNOWLEDGMENT This work was supported by National Institutes of Health Grant Number 1K08CA245483‐01. Myung Shin Sim was supported by NIH National Center for Advancing Translational Sciences (Grant 5UL1TR001881‐05). DATA AVAILABILITY STATEMENT Data are available on request from the authors.
AXICABTAGENE CILOLEUCEL, CYCLOPHOSPHAMIDE, FILGRASTIM, FLUDARABINE PHOSPHATE, TOCILIZUMAB
DrugsGivenReaction
CC BY-NC
33091961
18,449,659
2021-03-01
What was the dosage of drug 'CYCLOPHOSPHAMIDE'?
Filgrastim associations with CAR T-cell therapy. Little is known about the benefits and risks of myeloid growth factor administration after chimeric antigen receptor (CAR) T-cell therapy for diffuse large B-cell lymphoma (DLBCL). We present a retrospective analysis among 22 relapsed/refractory DLBCL patients who received CAR T-cell therapy with axicabtagene ciloleucel. Filgrastim was administered by physician discretion to seven patients (31.8%), and the median duration of neutropenia after lymphodepleting therapy was significantly shorter for those patients who received filgrastim (5 vs 15 days, P = .016). Five patients (22.7%) developed infection in the 30 days post-CAR T-cell therapy with three patients being Grade 3 or higher. There was no difference in the incidence and severity of infection based on filgrastim use (P = .274, P = .138). Among the seven patients that received filgrastim, six patients (85.7%) and four patients (57.1%) had evidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), respectively. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) and 7 patients (46.7%) had evidence of CRS and ICANS, respectively. There was no significant difference in the incidence of developing CRS or ICANS between the group of patients that received filgrastim and those that did not (P = .193, P = .647). However, there was a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042). Filgrastim administration after CAR T-cell therapy may lead to an increase in severity of CRS without decreasing infection rates. Abbreviations ANCabsolute neutrophil count ASTCTAmerican Society of Transplantation and Cellular Therapy CARchimeric antigen receptor CTCAECommon Terminology Criteria for Adverse Events DLBCLdiffuse large B‐cell lymphoma G‐CSFgranulocyte colony‐stimulating factor GM‐CSFgranulocyte‐macrophage colony‐stimulating factor ICANSimmune effector cell‐associated neurotoxicity syndrome IFNinterferon ILinterleukin IQRinterquartile range RICErituximab/ifosfamide/carboplatin/etoposide 1 INTRODUCTION While chimeric antigen receptor (CAR) T‐cell therapy is an exciting advancement in the treatment of patients with relapsed/refractory diffuse large B‐cell lymphoma (DLBCL), its application is limited by associated toxicities, including cytokine release syndrome (CRS), neurotoxicity and severe cytopenias, with 78% of patients developing Grade 3 or higher neutropenia in the ZUMA‐1 trial. 1 Recombinant granulocyte colony‐stimulating factor (G‐CSF) has been widely used to shorten the duration of neutropenia and risk of infection in other settings, such as neutropenic complications after conditioning for hematopoietic cell transplant. 2 However, in CAR T‐cell therapy, myeloid growth factors have the potential to increase the incidence and/or severity of CRS and immune effector cell‐associated neurotoxicity syndrome (ICANS) by promotion of proinflammatory cytokine secretion from monocytes and macrophages. 3 , 4 , 5 Preclinically, monocytes and macrophages are the main source of interleukin (IL)‐1 and IL‐6 during CRS. 4 , 5 Surprisingly, CRS severity is mediated not by CAR T‐cell‐derived cytokines, but by those of monocytes and macrophages. 4 , 5 Therefore, we sought to determine if a relationship exists between filgrastim administration, which stimulates myelopoiesis and enhances granulocyte function, and CRS. There is currently a lack of evidence to guide clinicians on the benefits and risks of recombinant myeloid growth factor use in CAR T‐cell treatment. 2 MATERIALS AND METHODS Between March 2018 and May 2019, we reviewed 22 patients with DLBCL treated with axicabtagene ciloleucel with or without concurrent use of filgrastim. Prior to CAR T‐cell infusion, all patients received standard lymphodepleting therapy with fludarabine 30 mg/m2/day and cyclophosphamide 500 mg/m2/day on days −5 through −3, except for one patient that received reduced doses for chronic kidney disease Stage IV (fludarabine 15 mg/m2/day and cyclophosphamide 375 mg/m2/day). Prophylactic tocilizumab 8 mg/kg was given to all patients at 36 hours after CAR T‐cell infusion, with additional doses of tocilizumab and/or steroids given for evidence of CRS/ICANS based on the American Society for Transplantation and Cellular Therapy (ASTCT) consensus grading system. 6 Filgrastim was administered at physician discretion after CAR T‐cell infusion at a weight‐based dose of either 300 or 480 mcg, and cumulative filgrastim dose was recorded within the first 30 days. Toxicity after CAR T‐cell therapy was also assessed up to 30 days postinfusion. Any patient with neutropenic fever was treated with broad‐spectrum antibiotics; no prophylactic antibiotics were given. Documented infections were graded according to Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. 7 Antibiotics were also administered for any confirmed infections. Chi‐squared test or Cochran‐Armitage test were performed to examine association between cohort characteristics and the study variable of administration with or without filgrastim. 3 RESULTS Baseline patient characteristics are displayed in Table 1. The median age at the time of CAR T‐cell therapy was 65.0 years (interquartile range [IQR], 57.0‐68.8). The majority (14 patients, 63.6%) had high‐grade DLBCL, while 5 patients had DLBCL not otherwise specified, 2 patients had transformed follicular lymphoma and 1 patient had Richter's transformation. Seven patients (31.8%) had refractory DLBCL, while 15 patients (68.2%) had relapsed DLBCL with a median number of relapses prior to CAR T‐cell therapy of 1.0 (IQR, 0.0‐2.0). Thirteen patients (59.1%) had received greater than or equal to three prior therapies. Only one patient (4.6%) had received autologous hematopoietic cell transplant prior to CAR T‐cell therapy. Ten patients (45.5%) received bridging therapy for high disease burden before CAR T‐cell therapy, which included rituximab/gemcitabine‐oxaliplatin (7 patients), cytarabine/thiotepa (1 patient), rituximab/dexamethasone/cytarabine/cisplatin (1 patient), rituximab/ifosfamide/carboplatin/etoposide (RICE, 1 patient) and rituximab/cyclophosphamide/dexamethasone (1 patient). The number of cycles of bridging therapy ranged from 1 to 3 with a median of 2. TABLE 1 Baseline patient and disease characteristics All patients (N = 22) Age at time of CAR T‐cell therapy (median, IQR) 65.0 (57.0, 68.8) ≥65 12 (54.6%) <65 10 (45.5%) Gender Male 12 (54.6%) Female 10 (45.5%) Ethnicity Caucasian 12 (54.6%) Hispanic 7 (31.8%) Asian 0 (0%) African American 1 (4.6%) Other 2 (9.1%) Disease type DLBCL NOS 5 (22.7%) Transformed follicular 2 (9.1%) Richter's transformation 1 (4.6%) High‐grade DLBCL 14 (63.6%) Cell of origin GCB 10 (45.5%) Non‐GCB 10 (45.5%) Unspecified 2 (9.1%) Relapse or Refractory Relapse 15 (68.2%) Refractory 7 (31.8%) Number of prior therapies <3 9 (40.9%) ≥3 13 (59.1%) ASCT prior to CAR T‐cell therapy 1 (4.6%) ECOG at time of CAR T‐cell therapy 0‐1 21 (95.5%) 2‐4 1 (4.6%) Bridging therapy given before CAR T‐cell therapy Yes 10 (45.5%) No 12 (54.6%) Abbreviations: ASCT, autologous stem cell transplant; CAR, chimeric antigen receptor; DLBCL, diffuse large B‐cell lymphoma; ECOG, Eastern Cooperative Oncology Group; GCB, germinal center B‐cell; NOS, not otherwise specified. Seven of the 22 patients (31.8%) received filgrastim by physician discretion at a target dose of 5 mcg/kg/day with 3 patients receiving 300 mcg/day and 4 patients receiving 480 mcg/day. The median start day of filgrastim was 2 days post‐CAR T‐cell therapy (range Day −2 to Day +7), and the median number of filgrastim doses was 6 (range, 3‐18). The median duration of neutropenia after CAR T‐cell therapy was 5 days (IQR, 4.5‐8.5) for patients who received filgrastim compared to 15 days (IQR, 8.0‐30.0) for patients who did not receive filgrastim (P = .016; Table 2). Fourteen patients (63.6%) developed neutropenic fever after CAR T‐cell therapy, 6 of whom received filgrastim and 8 of whom had not (P = .193; Table 2). Five patients (22.7%) developed an infection in the 30 days post‐CAR T‐cell therapy including Clostridium difficile colitis (one patient, Grade 3), Enterococcus faecalis bacteremia (one patient, Grade 4), pneumonia (one patient, Grade 3) and Herpes simplex virus (two patients, Grades 1 and 2). Among the seven patients that received filgrastim, three patients (42.9%) developed an infection and two patients (28.6%) developed a Grade 3 or greater infection. Among the 15 patients that did not receive filgrastim, 2 patients (13.2%) developed an infection and 1 patient (6.7%) developed a Grade 3 or greater infection. There was no difference in the incidence and severity of infection between patients who received filgrastim and those that did not (P = .274, P = .138; Table 2; Figure 1). TABLE 2 CAR T‐cell associated toxicity All patients (N = 22) G‐CSF administered (N = 7) G‐CSF not administered (N = 15) P value Median duration of neutropenia (days) (IQR) 10 (6.0, 25.8) 5 (4.5, 8.5) 15 (8.0, 30.0) .016 Febrile neutropenia No 8 (36.4%) 1 (14.3%) 7 (46.7%) .193 Yes 14 (63.6%) 6 (85.7%) 8 (53.5%) New infection .274 None 17 (77.3%) 4 (57.1%) 13 (86.7%) Any grade 5 (22.7%) 3 (42.9%) 2 (13.2%) CRS .193 None 8 (36.4%) 1 (14.3%) 7 (46.7%) Any grade 14 (63.6%) 6 (85.7%) 8 (53.3%) ICANS .647 None 11 (50.0%) 3 (42.9%) 8 (53.3%) Any grade 11 (50.0%) 4 (57.1%) 7 (46.7%) Steroids given .648 No 9 (40.9%) 2 (28.6%) 7 (46.7%) Yes 13 (59.1%) 5 (71.4%) 8 (53.3%) More than one dose of tocilizumab given .074 No 10 (45.5%) 1 (14.3%) 9 (60.0%) Yes 12 (54.6%) 6 (85.7%) 6 (40.0%) Abbreviations: CAR, chimeric antigen receptor; CRS, cytokine release syndrome; G‐CSF, granulocyte colony‐stimulating factor; ICANS, immune effector cell‐associated neurotoxicity syndrome. FIGURE 1 Severity of chimeric antigen receptor (CAR) T‐cell associated toxicities based on granulocyte colony‐stimulating factor (G‐CSF) use. ICANS, immune effector cell‐associated neurotoxicity syndrome [Color figure can be viewed at wileyonlinelibrary.com] CRS was noted in 14 patients overall (63.6%), and 4 patients (18.2%) had Grade 3 or higher CRS. Among the seven patients that received filgrastim, six patients (85.7%) had evidence of CRS and three patients (42.9%) had Grade 3 or higher CRS. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) had evidence of CRS and 1 patient (6.7%) had evidence of Grade 3 or higher CRS. ICANS was noted in 11 patients overall (50.0%), and 9 patients (40.9%) had Grade 3 or higher ICANS. Among the seven patients that received filgrastim, four patients (57.1%) had evidence of ICANS and three patients (42.9%) had evidence of Grade 3 or higher ICANS. Among the 15 patients that did not receive filgrastim, 7 patients (46.7%) had evidence of ICANS and 6 patients (40.0%) had Grade 3 or higher ICANS. There was no significant difference in the incidence of developing CRS (any grade) or ICANS (any grade) between the group of patients that received filgrastim and those that did not (P = .193, P = .647; Table 2). There was, however, a significant increase in the severity of CRS for patients that received filgrastim compared to those that did not (P = .042), but no increase in the severity of ICANS based on filgrastim use (P = .660; Figure 1). Thirteen patients (59.1%) received corticosteroids after CAR T‐cell treatment at a median cumulative dosage of 666.7 mg prednisone equivalents (IQR, 445.0‐933.3), with the majority (9 patients, 69.2%) receiving steroids in the first 5 days post‐CAR T‐cell infusion. Half of the patient cohort (12 patients, 54.6%) required at least one dose of tocilizumab in addition to the scheduled prophylactic dose. Among the seven patients that received filgrastim, 5 patients (71.4%) received corticosteroids and 6 patients (85.7%) received more than one dose of tocilizumab. Among the 15 patients that did not receive filgrastim, 8 patients (53.3%) received corticosteroids and 6 patients (40.0%) received more than one dose of tocilizumab. There was no association between filgrastim use and steroid use or administration of additional doses of tocilizumab (P = .648, P = .074). 4 DISCUSSION At our institution, filgrastim was administered after CAR T‐cell therapy at physician discretion with less than half of our patient cohort receiving filgrastim (seven patients, 31.8%). Importantly, although the median duration of neutropenia was significantly lower for patients who received filgrastim, there was no difference in the development or severity of infections (P = .274, P = .138). Instead, while patients given filgrastim were not more likely to develop CRS (P = .193), the severity of CRS was higher in those that received filgrastim (P = .042), as has been suggested by preclinical data modeling myeloid growth factors in combination with CAR T‐cells. 4 , 5 The development of CRS is related to activation of in vivo T‐cell expansion and the production of T‐cell effector cytokines, including IL‐6, IL‐10 and interferon (IFN)‐γ, 8 which are also downstream products of myeloid cells stimulated by filgrastim. One preclinical study showed that higher levels of murine G‐CSF correlated strongly with CRS severity and survival. 5 Elevated systemic levels of IL‐6 in particular have been associated with severe CRS, and IL‐6 receptor blockade with the monoclonal antibody tocilizumab is an important agent used to reduce CRS toxicity. 9 , 10 Of all the cytokines analyzed in the ZUMA‐2 study, only peak levels of granzyme B and granulocyte‐macrophage colony stimulating factor (GM‐CSF, a closely related protein to G‐CSF) were associated with severe CRS and severe ICANS. 11 CAR T‐cell associated neurotoxicity has been associated with elevations in similar inflammatory markers to CRS (IL‐6, IL‐10, IFNγ), in addition to higher serum levels of G‐CSF and GM‐CSF. 12 , 13 Mouse models of CRS and ICANS have suggested that the main driver of CAR T‐cell neurotoxicity is actually IL‐1 secretion from activated macrophages. 4 , 5 Unlike its efficacy in CRS, tocilizumab did not protect mice from lethal neurotoxicity, a finding analogous in humans; however the IL‐1 receptor antagonist anakinra abolished both CRS and neurotoxicity. 4 , 12 Interestingly, in addition to its stimulatory effects on granulocytes, filgrastim has immunomodulatory effects on other immune cells and can inhibit inflammatory cytokine production within monocytes or macrophages. 14 This may account for why we found no association with filgrastim and the development or severity of neurotoxicity (P = .647, P = .660). GM‐CSF, which induces both granulocytes and macrophages, may be more closely tied to neurotoxic effects and, in fact, GM‐CSF inhibition has been shown to reduce neuroinflammation and prevent CRS. 15 The guidelines on use of filgrastim and other growth factors with CAR T‐cell therapy are not standardized. While some recommend administration of myeloid growth factors once absolute neutrophil count (ANC) decreases to <500/μL and to continue until ANC increases to ≥1500/μL, 16 others recommend administration of filgrastim only if patients develop Grade 1 CRS at the same time as neutropenia or in the setting of neutropenic fever. 17 The use of myeloid growth factors across institutions employing CAR T‐cell therapy is also mixed. In an electronic survey on the current administrative, logistic and toxicity management practices of CAR T‐cell therapy across the United States by the ASTCT, out of 28 respondents, 46% used growth factor if allowed by product labeling, 29% never administered growth factor, 14% determined the use of growth factor on a patient‐specific basis and 11% administered growth factor to all patients. 18 Although our study is limited by its small sample size and retrospective nature, we suggest that myeloid growth factor administration be used with caution in patients undergoing CAR T‐cell therapy. We found an association between filgrastim use and CRS severity, suggesting that increasing the number of patients in the study would strengthen the association. However, the effect size of the association was small. Further studies are required to determine the safety of myeloid growth factors after CAR T‐cell therapy. CONFLICT OF INTEREST The authors declared no potential conflicts of interest. ETHICS STATEMENT This study was approved by the University of California Los Angeles Institutional Review Board. Informed consent was not required for this study. ACKNOWLEDGMENT This work was supported by National Institutes of Health Grant Number 1K08CA245483‐01. Myung Shin Sim was supported by NIH National Center for Advancing Translational Sciences (Grant 5UL1TR001881‐05). DATA AVAILABILITY STATEMENT Data are available on request from the authors.
500 MILLIGRAM/SQ. METER, QD
DrugDosageText
CC BY-NC
33091961
18,449,659
2021-03-01