Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 363
Filter
1.
BMJ Case Rep ; 17(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38697679

ABSTRACT

Ornithine transcarbamylase deficiency (OTCD) is a rare, X linked disorder that can manifest in late adulthood in heterozygous females as severe hyperammonaemia following environmental stressors. We present a case of hyperammonaemic encephalopathy that was triggered by glucocorticoid administration in an adult woman with heterozygous OTCD with clinical response to haemodialysis, ammonia scavengers and a high-calorie, low-protein diet.


Subject(s)
Hyperammonemia , Ornithine Carbamoyltransferase Deficiency Disease , Humans , Female , Ornithine Carbamoyltransferase Deficiency Disease/complications , Ornithine Carbamoyltransferase Deficiency Disease/diagnosis , Hyperammonemia/chemically induced , Glucocorticoids/therapeutic use , Glucocorticoids/adverse effects , Renal Dialysis , Brain Diseases/chemically induced , Brain Diseases/etiology , Middle Aged , Diet, Protein-Restricted/adverse effects
2.
Biomolecules ; 14(3)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38540788

ABSTRACT

(1) Background: Valproic acid (VPA) is one of the frequently prescribed antiepileptic drugs and is generally considered well tolerated. However, VPA neurologic adverse effects in the absence of liver failure are fairly common, suggesting that in the mechanism for the development of VPA-induced encephalopathy, much more is involved than merely the exposure to hyperammonemia (HA) caused by liver insufficiency to perform detoxification. Taking into account the importance of the relationship between an impaired brain energy metabolism and elevated ammonia production, and based on the ability of VPA to interfere with neuronal oxidative pathways, the current study intended to investigate a potential regional ammoniagenic effect of VPA on rats' brains by determining activities of the enzymes responsible for ammonia production and neutralization. (2) Methods: Rats received a single intraperitoneal injection of VPA (50, 100, 250, 500 mg/kg). Plasma, the neocortex, the cerebellum, and the hippocampus were collected at 30 min after injection. The levels of ammonia, urea, aspartate aminotransferase (AST), and alanine aminotransferase (ALT) were measured in blood plasma. The activities of glutaminase and glutamate dehydrogenase (GDH) in mitochondria and the activities of AMP deaminase (AMPD), adenosine deaminase (ADA), and glutamine synthetase (GS) in cytosolic fractions isolated from rat brain regions were measured. Ammonia, ALT, and AST values were determined in the mitochondrial and cytosolic fractions. (3) Results: Multi-dose VPA treatment did not significantly affect the plasma levels of ammonia and urea or the ALT and AST liver enzymes. Significant dose-independent increases in the accumulation of ammonia were found only in the cytosol from the cerebellum and there was a strong correlation between the ammonia level and the ADA activity in this brain structure. A significant decrease in the AMPD and AST activities was observed, while the ALT activity was unaffected. Only the highest VPA dose (500 mg/kg) was associated with significantly less activity of GS compared to the control in all studied brain structures. In the mitochondria of all studied brain structures, VPA caused a dose-independent increases in ammonia levels, a high concentration of which was strongly and positively correlated with the increased GDH and ALT activity, while glutaminase activity remained unchanged, and AST activity significantly decreased compared to the control in all studied brain structures. (4) Conclusions: This study highlights the rat brain region-specific ammoniagenic effects of VPA, which may manifest themselves in the absence of hyperammonemia. Further research should analyze how the responsiveness of the different brain regions may vary in VPA-treated animals that exhibit compromised energy metabolism, leading to increased ammoniagenesis.


Subject(s)
Hyperammonemia , Valproic Acid , Rats , Animals , Valproic Acid/adverse effects , Glutaminase , Hyperammonemia/chemically induced , Hyperammonemia/metabolism , Ammonia/metabolism , Urea
3.
Bipolar Disord ; 26(2): 200-203, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38253983

ABSTRACT

OBJECTIVE: Treatment of bipolar disorder (BD) involves complexities especially when patients come with significant sensitivity to various psychotropic medications and comorbidities. The following cases aim to recapitulate and discuss some of such situations. CASES: Case 1: A 36-year-old man with intellectual development disorder and BD experienced catatonia, seizures, and hyperammonemia following valproate administration. Treatment involved electroconvulsive therapy (ECT) and a tailored medication regimen, ultimately leading to stability. Case 2: A 63-year-old man with long-standing BD exhibited resistance to lithium and valproate of late, having co-existing essential tremors and cerebellar atrophy. Multiple medication trials led to side effects, requiring ECT for symptom improvement, followed by a carefully adjusted maintenance regimen. CONCLUSION: Medication side effects can pose major challenges in treatment of BD. Comprehensive evaluation and monitoring are essential. ECT can prove valuable in such cases. There is pressing need to develop more safer treatment alternatives, especially considering the progressively ageing society.


Subject(s)
Antipsychotic Agents , Bipolar Disorder , Hyperammonemia , Male , Humans , Adult , Middle Aged , Bipolar Disorder/diagnosis , Valproic Acid/adverse effects , Antipsychotic Agents/therapeutic use , Hyperammonemia/chemically induced , Hyperammonemia/therapy , Hyperammonemia/complications , Atrophy/chemically induced , Atrophy/complications , Atrophy/drug therapy
4.
J Oncol Pharm Pract ; 30(3): 576-583, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38258317

ABSTRACT

OBJECTIVE: To review the evidence of uncommon but fatal adverse event of hyperammonemic encephalopathy by tyrosine kinase inhibitors (TKI) and the possible mechanisms underlying this condition and to describe the case of a patient that developed drug-induced hyperammonemic encephalopathy related to TKI. DATA SOURCES: Literature search of different databases was performed for studies published from 1 January 1992 to 7 May 2023. The search terms utilized were hyperammonemic encephalopathy, TKI, apatinib, pazopanib, sunitinib, imatinib, sorafenib, regorafenib, trametinib, urea cycle regulation, sorafenib, carbamoyl-phosphate synthetase 1, ornithine transcarbamylase, argininosuccinate synthetase, argininosuccinate lyase, arginase 1, Mitogen activated protein kinases (MAPK) pathway and mTOR pathway, were used individually search or combined. DATA SUMMARY: Thirty-seven articles were included. The articles primarily focused in hyperammonemic encephalopathy case reports, management of hyperammonemic encephalopathy, urea cycle regulation, autophagy, mTOR and MAPK pathways, and TKI. CONCLUSION: Eighteen cases of hyperammonemic encephalopathy were reported in the literature from various multitargeted TKI. The mechanism of this event is not well-understood but some authors have hypothesized vascular causes since some of TKI are antiangiogenic, however our literature review shows a possible relationship between the urea cycle and the molecular inhibition exerted by TKI. More preclinical evidence is required to unveil the biochemical mechanisms responsible involved in this process and clinical studies are necessary to shed light on the prevalence, risk factors, management and prevention of this adverse event. It is important to monitor neurological symptoms and to measure ammonia levels when manifestations are detected.


Subject(s)
Hyperammonemia , Protein Kinase Inhibitors , Humans , Hyperammonemia/chemically induced , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Male , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Brain Diseases/chemically induced
7.
BMJ Case Rep ; 16(11)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38035682

ABSTRACT

There is limited information about sodium valproate-induced hyperammonaemia encephalopathy (VPAIHE). The aim of this case report is to provide medical practitioners with a greater awareness of the possible development of hyperammonaemia due to sodium valproate use and its associated complications.This paper describes a middle-aged man with a history of bipolar affective disorder who was admitted with a manic relapse secondary to medication non-compliance. His admission was complicated by an intensive care unit admission to manage medical compromise in the context of sodium VPAIHE.


Subject(s)
Bipolar Disorder , Brain Diseases , Hyperammonemia , Male , Middle Aged , Humans , Valproic Acid/adverse effects , Hyperammonemia/chemically induced , Hyperammonemia/drug therapy , Neoplasm Recurrence, Local/drug therapy , Bipolar Disorder/drug therapy , Bipolar Disorder/psychology , Brain Diseases/chemically induced , Brain Diseases/drug therapy
8.
Ann Biol Clin (Paris) ; 81(4): 365-377, 2023 10 20.
Article in French | MEDLINE | ID: mdl-37864442

ABSTRACT

Asparaginase is a key molecule in the treatment of acute lymphoblastic leukemia. It has improved response rates to chemotherapy. However, this is not without consequences. Therapeutic efficacy is sometimes achieved at the expense of toxicities that can lead to treatment discontinuation. Among them, patients can develop hyperammonemia which can sometimes be symptomatic leading to neurological disorders that can go as far as hyperammonemic coma or even death. Through a review of the current state of the literature, the objective is to understand the disparity of ammonia values as well as the clinical heterogeneity for a given ammonia concentration. A review of the literature including more than eighty publications was performed. The glutaminase activity of asparaginase seems to play an important role in the development of hyperammonia. At present, no risk factors have been identified for the development of hyperammonemia. On the other hand, the question of the impact of pre-analysis phase arises. Indeed, asparaginase continues to exert its activity in vitro, which leads to an artefactual increase in ammonia.


Subject(s)
Antineoplastic Agents , Hyperammonemia , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Asparaginase/adverse effects , Ammonia/therapeutic use , Hyperammonemia/chemically induced , Hyperammonemia/diagnosis , Hyperammonemia/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Risk Factors , Antineoplastic Agents/adverse effects
9.
Clin Neurol Neurosurg ; 233: 107962, 2023 10.
Article in English | MEDLINE | ID: mdl-37717359

ABSTRACT

BACKGROUND: Hyperammonemia can occur after acute overdose or chronic use of valproic acid (VPA). Although VPA-related hyperammonemic encephalopathy (VHE) is a rare complication of VPA therapy, early recognition of VHE and identifying its risk factors are important because VHE can lead to loss of consciousness and increased seizure frequency. PURPOSE: The purpose of our study is to evaluate the risk factors of hyperammonemia in epilepsy patients during treatment with VPA therapy. METHODS: We reviewed the medical records of 1084 adult patients with epilepsy and enrolled 116 patients with VPA therapy who had results of blood levels of ammonia over a 3-year period. Hyperammonemia was defined as a blood ammonia level exceeding 80 µg/dL. Correlations of blood levels of ammonia with dosages and blood levels of VPA were evaluated. We further performed univariate and multivariate linear regression analyses to identify risk factors for hyperammonemia in epilepsy patients treated with VPA therapy. RESULTS: Blood levels of ammonia were well correlated with dosages of VPA (p = 0.036), but not with blood levels of VPA (p = 0.463). Hyperammonemia was more common in patients with higher VPA dosage and higher total drug loads of concurrent antiseizure medications (ASMs). Hyperammonemia was also associated with the use of topiramate and phenobarbital. In multivariate analysis, we identified total drug load of ASMs (p = 0.003) and use of topiramate (p = 0.007) as independent predictors of hyperammonemia. Four patients (4/116, 3.4 %) had clinical symptoms of VHE. Three of them had hyperammonemia while the other patient had normal blood level of ammonia with a high blood level of VPA. CONCLUSION: Our study shows that higher total drug loads of concurrent ASMs and use of topiramate were independent risk factors of hyperammonemia in epilepsy patients with VPA therapy. Although the incidence of VHE was not high in our study, clinicians should be aware of this potential adverse effect of VPA therapy, especially in patients with polytherapy of ASMs including topiramate.


Subject(s)
Epilepsy , Hyperammonemia , Adult , Humans , Valproic Acid/adverse effects , Anticonvulsants/adverse effects , Topiramate/adverse effects , Hyperammonemia/chemically induced , Ammonia/therapeutic use , Epilepsy/drug therapy , Risk Factors
11.
Acta Biomater ; 168: 515-528, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37433359

ABSTRACT

l-Ornithine (Orn) is a core amino acid responsible for ammonia detoxification in the body via the hepatic urea cycle. Clinical studies in Orn therapy have focused on interventions for hyperammonemia-associated diseases, such as hepatic encephalopathy (HE), a life-threatening neurological symptom affecting more than 80% of patients with liver cirrhosis. However, its low molecular weight (LMW) causes Orn to diffuse nonspecifically and be rapidly eliminated from the body after oral administration, resulting in unfavorable therapeutic efficacy. Hence, Orn is constantly supplied by intravenous infusion in many clinical settings; however, this treatment inevitably decreases patient compliance and limits its application in long-term management. To improve the performance of Orn, we designed self-assembling polyOrn-based nanoparticles for oral administration through ring-opening polymerization of Orn-N-carboxy anhydride initiated with amino-ended poly(ethylene glycol), followed by acylation of free amino groups in the main chain of the polyOrn segment. The obtained amphiphilic block copolymers, poly(ethylene glycol)-block-polyOrn(acyl) (PEG-block-POrn(acyl)), enabled the formation of stable nanoparticles (NanoOrn(acyl)) in aqueous media. We employed the isobutyryl (iBu) group for acyl derivatization in this study (NanoOrn(iBu)). In the healthy mice, daily oral administration of NanoOrn(iBu) for one week did not induce any abnormalities. In the mice exhibiting acetaminophen (APAP)-induced acute liver injury, oral pretreatment with NanoOrn(iBu) effectively reduced systemic ammonia and transaminases levels compared to the LMW Orn and untreated groups. The results suggest that the application of NanoOrn(iBu) is of significant clinical value with the feasibility of oral delivery and improvement in APAP-induced hepatic pathogenesis. STATEMENT OF SIGNIFICANCE: Liver injury is often accompanied by hyperammonemia, a life-threatening condition characterized by elevated blood ammonia levels. Current clinical treatments for reducing ammonia typically entail the invasive approach of intravenous infusion, involving the administration of l-ornithine (Orn) or a combination of Orn and L-aspartate. This method is employed due to the poor pharmacokinetics associated with these compounds. In our pursuit of enhancing therapy, we have developed an orally administrable nanomedicine based on Orn-based self-assembling nanoparticle (NanoOrn(iBu)), which provides sustained Orn supply to the injured liver. Oral administration of NanoOrn(iBu) to healthy mice did not cause any toxic effects. In a mouse model of acetaminophen-induced acute liver injury, oral administration of NanoOrn(iBu) surpassed Orn in reducing systemic ammonia levels and liver damage, thereby establishing NanoOrn(iBu) as a safe and effective therapeutic option.


Subject(s)
Hyperammonemia , Mice , Animals , Hyperammonemia/chemically induced , Hyperammonemia/complications , Hyperammonemia/drug therapy , Ornithine/pharmacology , Ornithine/therapeutic use , Ornithine/metabolism , Acetaminophen/pharmacology , Polymers/pharmacology , Ammonia/metabolism , Ammonia/pharmacology , Nanomedicine , Liver , Polyethylene Glycols/pharmacology
12.
CNS Drugs ; 37(8): 725-731, 2023 08.
Article in English | MEDLINE | ID: mdl-37466895

ABSTRACT

BACKGROUND: Valproate-induced encephalopathy (VIE) affects between 0.1% and 2.5% of patients under long-term epilepsy treatment. Its frequency and characteristics in adults with status epilepticus (SE) is, however, unknown. OBJECTIVE: The aim of this study was to characterize the frequency and the clinico-biological characteristics of VIE in adult SE patients. METHODS: We reviewed all patients included in our institutional SE registry who were treated for an SE episode between November 2021 and February 2023 and identified 39 patients who received valproate for their SE treatment. Acute VIE was defined by worsening of consciousness having led to the discontinuation of valproate, and improvement of consciousness within 96 hours after discontinuation of valproate during acute hospital treatment. RESULTS: Patients had a mean valproate intravenous loading dose of 34.5 mg/kg and a mean maintenance dose of 15.3 mg/kg/d (1078 mg/d). Four out of 29 patients with measured ammonium had hyperammonemia. We identified four (10%) patients fulfilling acute VIE criteria. Median time from administration of valproate to the occurrence of VIE, and to resolution of VIE after cessation of valproate treatment, was 2 days for each. Three of the four VIE patients had no associated hyperammonemia. Patients who developed VIE more frequently had a history of liver disease (p = 0.023), and tended to be younger, but other clinical variables did not differ significantly from patients without VIE, including valproate loading or maintenance doses, SE cause, duration or severity, other concomitant antiseizure medications (none received topiramate, phenobarbital, or primidone). CONCLUSION: Pending larger studies, VIE in SE seems relatively frequent and difficult to foresee; clinical alertness to symptoms is mandatory, even without hyperammonemia, and valproate withdrawal should be considered in suspected cases.


Subject(s)
Brain Diseases , Hyperammonemia , Status Epilepticus , Adult , Humans , Anticonvulsants/adverse effects , Brain Diseases/chemically induced , Brain Diseases/drug therapy , Hyperammonemia/chemically induced , Hyperammonemia/drug therapy , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Valproic Acid/adverse effects
13.
Mol Genet Metab ; 139(3): 107627, 2023 07.
Article in English | MEDLINE | ID: mdl-37327713

ABSTRACT

Hyperammonemia has been reported following asparaginase administration, consistent with the mechanisms of asparaginase, which catabolizes asparagine to aspartic acid and ammonia, and secondarily converts glutamine to glutamate and ammonia. However, there are only a few reports on the treatment of these patients, which varies widely from watchful waiting to treatment with lactulose, protein restriction, sodium benzoate, and phenylbutyrate to dialysis. While many patients with reported asparaginase-induced hyperammonemia (AIH) are asymptomatic, some have severe complications and even fatal outcomes despite medical intervention. Here, we present a cohort of five pediatric patients with symptomatic AIH, which occurred after switching patients from polyethylene glycolated (PEG)- asparaginase to recombinant Crisantaspase Pseudomonas fluorescens (4 patients) or Erwinia (1 patient) asparaginase, and discuss their subsequent management, metabolic workup, and genetic testing. We developed an institutional management plan, which gradually evolved based on our local experience and previous treatment modalities. Because of the significant reduction in glutamine levels after asparaginase administration, sodium benzoate should be used as a first-line ammonia scavenger for symptomatic AIH instead of sodium phenylacetate or phenylbutyrate. This approach facilitated continuation of asparaginase doses, which is known to improve cancer outcomes. We also discuss the potential contribution of genetic modifiers to AIH. Our data highlights the need for increased awareness of symptomatic AIH, especially when an asparaginase with higher glutaminase activity is used, and its prompt management. The utility and efficacy of this management approach should be systematically investigated in a larger cohort of patients.


Subject(s)
Antineoplastic Agents , Hyperammonemia , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Child , Asparaginase/adverse effects , Phenylbutyrates/therapeutic use , Hyperammonemia/chemically induced , Hyperammonemia/drug therapy , Sodium Benzoate/adverse effects , Glutamine/adverse effects , Ammonia , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Treatment Outcome , Antineoplastic Agents/adverse effects
14.
J Clin Pathol ; 76(8): 501-509, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37164630

ABSTRACT

Hyperammonaemia (HA) as a consequence of numerous primary or secondary causes, gives rise to clinical manifestations due to its toxic effects on the brain. The neurological consequences broadly reflect the ammonia level, duration and age, with paediatric patients being more susceptible. Drug-induced HA may arise due to either decreased ammonia elimination or increased production. This is associated most frequently with use of valproate and presents a dilemma between ongoing therapeutic need, toxicity and the possibility of an alternative cause. As there is no specific test for drug-induced HA, prompt discussion with a metabolic physician is recommended, as the neurotoxic effects are time-dependent. Specific guidelines for managing drug-induced HA have yet to be published and hence the treatment approach outlined in this review reflects that outlined in relevant urea cycle disorder guidelines.


Subject(s)
Hyperammonemia , Humans , Child , Hyperammonemia/chemically induced , Hyperammonemia/diagnosis , Ammonia/metabolism , Brain/metabolism , Valproic Acid/adverse effects , Valproic Acid/metabolism
15.
Neurol Sci ; 44(10): 3669-3673, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37243793

ABSTRACT

BACKGROUND: Valproic acid (VPA) is a prevalent antiseizure medication (ASM) used to treat epilepsy. Valproate-related hyperammonemic encephalopathy (VHE) is a type of encephalopathy that can occur during neurocritical situations. In VHE, the electroencephalogram (EEG) displays diffuse slow waves or periodic waves, and there is no generalized suppression pattern. CASE PRESENTATION: We present a case of a 29-year-old female with a history of epilepsy who was admitted for convulsive status epilepticus (CSE), which was controlled by intravenous VPA, as well as oral VPA and phenytoin. The patient did not experience further convulsions but instead developed impaired consciousness. Continuous EEG monitoring revealed a generalized suppression pattern, and the patient was unresponsive. The patient's blood ammonia level was significantly elevated at 386.8 µmol/L, indicating VHE. Additionally, the patient's serum VPA level was 58.37 µg/ml (normal range: 50-100 µg/ml). After stopping VPA and phenytoin and transitioning to oxcarbazepine for anti-seizure and symptomatic treatment, the patient's EEG gradually returned to normal, and her consciousness was fully restored. DISCUSSION: VHE can cause the EEG to display a generalized suppression pattern. It is crucial to recognize this specific situation and not to infer a poor prognosis based on this EEG pattern.


Subject(s)
Brain Diseases , Epilepsy , Hyperammonemia , Neurotoxicity Syndromes , Humans , Female , Adult , Valproic Acid/adverse effects , Anticonvulsants/adverse effects , Phenytoin/adverse effects , Epilepsy/drug therapy , Epilepsy/complications , Brain Diseases/complications , Electroencephalography , Neurotoxicity Syndromes/drug therapy , Hyperammonemia/chemically induced , Hyperammonemia/drug therapy
16.
J Clin Psychopharmacol ; 43(3): 283-294, 2023.
Article in English | MEDLINE | ID: mdl-37126830

ABSTRACT

BACKGROUND: Hyperammonemia is an adverse effect that poses clinical uncertainty around valproic acid (VPA) use. The prevalence of symptomatic and asymptomatic hyperammonemia and its relationship to VPA concentration is not well established. There is also no clear guidance regarding its management. This results in variability in the monitoring and treatment of VPA-induced hyperammonemia. To inform clinical practice, this systematic review aims to summarize evidence available around VPA-associated hyperammonemia and its prevalence, clinical outcomes, and management. METHODS: An electronic search was performed through Ovid MEDLINE, Ovid Embase, Web of Science, and PsycINFO using search terms that identified hyperammonemia in patients receiving VPA. Two reviewers independently performed primary title and abstract screening with a third reviewer resolving conflicting screening results. This process was repeated during the full-text review process. RESULTS: A total of 240 articles were included. Prevalence of asymptomatic hyperammonemia (5%-73%) was higher than symptomatic hyperammonemia (0.7%-22.2%) and occurred within the therapeutic range of VPA serum concentration. Various risk factors were identified, including concomitant medications, liver injury, and defects in carnitine metabolism. With VPA discontinued, most symptomatic patients returned to baseline mental status with normalized ammonia level. There was insufficient data to support routine monitoring of ammonia level for VPA-associated hyperammonemia. CONCLUSIONS: Valproic acid-associated hyperammonemia is a common adverse effect that may occur within therapeutic range of VPA. Further studies are required to determine the benefit of routine ammonia level monitoring and to guide the management of VPA-associated hyperammonemia.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Epilepsy , Hyperammonemia , Humans , Valproic Acid/adverse effects , Anticonvulsants/adverse effects , Epilepsy/drug therapy , Hyperammonemia/chemically induced , Ammonia/adverse effects , Clinical Decision-Making , Uncertainty
17.
Cancer Chemother Pharmacol ; 92(1): 7-14, 2023 07.
Article in English | MEDLINE | ID: mdl-37204512

ABSTRACT

PURPOSE: Fluoropyrimidines are anticancer drugs and can cause hyperammonemia both intravenously and orally. Renal dysfunction may interact with fluoropyrimidine to cause hyperammonemia. We performed quantitative analyses of hyperammonemia using a spontaneous report database to examine the frequency of intravenously and orally administered fluoropyrimidine, the reported frequency of fluoropyrimidine-related regimens, and fluoropyrimidine's interactions with chronic kidney disease (CKD). METHODS: This study used data collected between April 2004 and March 2020 from the Japanese Adverse Drug Event Report database. The reporting odds ratio (ROR) of hyperammonemia was calculated for each fluoropyrimidine drug and was adjusted for age and sex. Heatmaps depicting the use of anticancer agents in patients with hyperammonemia were drawn. The interactions between CKD and the fluoropyrimidines were also calculated. These analyses were performed using multiple logistic regression. RESULTS: Hyperammonemia was observed in 861 of the 641,736 adverse events reports. Fluorouracil was the most frequent drug associated with hyperammonemia (389 cases). The ROR of hyperammonemia was 32.5 (95% CI 28.3-37.2) for intravenously administered fluorouracil, 4.7 (95% CI 3.3-6.6) for orally administered capecitabine, 1.9 (95% CI 0.87-4.3) for tegafur/uracil, and 2.2 (95% CI 1.5-3.2) for orally administered tegafur/gimeracil/oteracil. Calcium levofolinate, oxaliplatin, bevacizumab, and irinotecan were the most frequently reported agents in cases of hyperammonemia with intravenously administered fluorouracil. The coefficient of the interaction term between CKD and fluoropyrimidines was 1.12 (95% CI 1.09-1.16). CONCLUSION: Hyperammonemia cases were more likely to be reported with intravenous fluorouracil than orally administered fluoropyrimidines. Fluoropyrimidines might interact with CKD in hyperammonemia cases.


Subject(s)
Antineoplastic Agents , Drug-Related Side Effects and Adverse Reactions , Hyperammonemia , Humans , Antimetabolites , Antineoplastic Agents/adverse effects , Capecitabine , Drug-Related Side Effects and Adverse Reactions/drug therapy , Fluorouracil , Hyperammonemia/chemically induced , Tegafur , Japan
18.
Gan To Kagaku Ryoho ; 50(4): 493-495, 2023 Apr.
Article in Japanese | MEDLINE | ID: mdl-37066465

ABSTRACT

Hyperammonemia induced by 5-fluorouracil(5-FU)is known as a rare adverse event, but there are few reports of hyperammonemia occurring during FP(5-FU plus CDDP)treatment for esophageal cancer. We report a case of esophageal cancer with consciousness disorder due to hyperammonemia during FP treatment with an examination of some of the relevant literature. The patient was a man of approximately 70 years of age who was received FP treatment. He showed consciousness disorder on day 4. A blood test showed hyperammonemia(427µg/dL), which was considered to be the cause of his consciousness disorder. He was treated with branched chain amino acid infusion, lactulose and kanamycin and made a full recovery. An operation for esophageal cancer was performed after 3 months and he is currently followed up without recurrence. Hyperammonemia should be considered as a differential diagnosis of consciousness disorder during chemotherapy including 5-FU.


Subject(s)
Esophageal Neoplasms , Hyperammonemia , Male , Humans , Hyperammonemia/chemically induced , Hyperammonemia/drug therapy , Consciousness Disorders/chemically induced , Fluorouracil , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/etiology , Antineoplastic Combined Chemotherapy Protocols/adverse effects
19.
BMC Psychiatry ; 23(1): 276, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37081388

ABSTRACT

BACKGROUND: Methamphetamine is an addictive drug with various effects on the neurotransmitters in the central nervous system. Methamphetamine-induced encephalopathy in the absence of hyperammonemia presents a unique challenge in a clinical setting. Previously published cases of methamphetamine-induced encephalopathy suggested that methamphetamine-induced hepatotoxicity and subsequent hyperammonemia may be the cause of encephalopathy. However, the literature is limited on methamphetamine-induced encephalopathy without hyperammonemia. CASE: This case presents a disoriented patient with methamphetamine use disorder in acute toxicity, unable to ambulate independently, and poorly responsive to verbal stimuli. The patient was found to have normal ammonia levels. DISCUSSION: This patient's presentation and laboratory findings, namely normal ammonia levels, suggest a different pathophysiological pathway for methamphetamine-induced encephalopathy. One potential pathway is through the direct action of methamphetamine on the central nervous system through acute disruption of neurotransmitter signaling and disruption of the blood-brain barrier. CONCLUSION: Further research should be conducted into the prevalence and pathophysiology of methamphetamine-induced encephalopathy in the absence of hyperammonemia. KEY POINTS: Methamphetamine-induced encephalopathy (MIE) in the absence of hyperammonemia presents a unique challenge in a clinical setting. Previously published cases of MIE suggest that methamphetamine-induced hepatotoxicity and subsequent hyperammonemia may be the cause of encephalopathy. Further research should be conducted into the prevalence and pathophysiology of MIE in the absence of hyperammonemia.


Subject(s)
Brain Diseases , Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Hyperammonemia , Methamphetamine , Humans , Hyperammonemia/chemically induced , Methamphetamine/adverse effects , Ammonia/adverse effects , Ammonia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...