Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Int J Infect Dis ; 143: 107021, 2024 Jun.
Article En | MEDLINE | ID: mdl-38561040

OBJECTIVES: Evaluate and compare the efficacy and safety of molnupiravir and favipiravir in outpatients with mild to moderate COVID-19 and at risk of severe COVID-19. METHODS: In an open-label, parallel-group, multicenter trial in Thailand, participants with moderate COVID-19 and at least one factor associated with severe COVID-19 were randomly assigned 1:1 to receive oral molnupiravir or oral favipiravir (standard of care). Phone calls for remote symptom assessment were made on Days 6, 15, and 29. Participants with worsening symptoms were instructed to return to the hospital. The primary endpoint was pulmonary involvement by Day 29, as evidenced by ≥2 of the following: dyspnea, oxygen saturation <92% or imaging. RESULTS: Nine hundred seventy-seven participants (487 molnupiravir, 490 favipiravir) were enrolled from 8 July 2022 to 19 January 2023. 98% had received ≥1 dose of COVID-19 vaccine and 83% ≥3 doses. By Day 29, pulmonary involvement occurred in 0% (0/483) in molnupiravir arm versus 1% (5/482) in favipiravir arm (-1.0%; Newcombe 95.2% CI: -2.4% to -0.0%; P = 0.021); all-cause death in 0% (0/483) and <1% (1/482); COVID-19 related hospitalization in <1% (1/483) and 1% (3/482); treatment-related adverse event in 1% (5/483) and 1% (4/486); and serious adverse event in 1% (4/483) and 1% (4/486). CONCLUSIONS: Favipiravir and molnupiravir had a similar efficacy and safety profile. Whether either of the two reduced the risk of complications during the omicron era in this population with a low risk of pulmonary involvement and a high vaccine coverage remains unclear. There were no differences in any of the safety endpoints. THAI CLINICAL TRIALS REGISTRY ID: TCTR20230111009.


Amides , Antiviral Agents , COVID-19 Drug Treatment , Cytidine/analogs & derivatives , Pyrazines , SARS-CoV-2 , Humans , Amides/therapeutic use , Male , Pyrazines/therapeutic use , Pyrazines/adverse effects , Pyrazines/administration & dosage , Female , Thailand , Antiviral Agents/therapeutic use , Antiviral Agents/adverse effects , Antiviral Agents/administration & dosage , Middle Aged , Adult , Cytidine/therapeutic use , Cytidine/adverse effects , Cytidine/administration & dosage , Hydroxylamines/therapeutic use , Hydroxylamines/adverse effects , Hydroxylamines/administration & dosage , Aged , Treatment Outcome , COVID-19 , Outpatients
3.
Ann Intern Med ; 175(4): JC40, 2022 04.
Article En | MEDLINE | ID: mdl-35377719

SOURCE CITATION: Jayk Bernal A, Gomes da Silva MM, Musungaie DB, et al. Molnupiravir for oral treatment of Covid-19 in nonhospitalized patients. N Engl J Med. 2022;386:509-20. 34914868.


COVID-19 , Adult , Cytidine/analogs & derivatives , Hospitalization , Humans , Hydroxylamines/adverse effects , SARS-CoV-2
6.
Diabetes Metab Syndr ; 16(2): 102396, 2022 Feb.
Article En | MEDLINE | ID: mdl-35051686

BACKGROUND AND AIMS: Molnupiravir is a newer oral antiviral drug that has recently received emergency use authorization (EUA) in USA, UK and India. We aim to conduct an update on our previous systematic review to provide practical clinical guideline for using molnupiravir in patients with COVID-19. METHODS: We systematically searched the electronic database of PubMed, MedRxiv and Google Scholar until January 5, 2022, using key MeSH keywords. RESULTS: Final result of phase 3 study in 1433 non-hospitalized COVID-19 patients showed a significant reduction in composite risk of hospital admission or death (absolute risk difference, -3.0% [95% confidence interval {CI}, -5.9 to -0.1%]; 1-sided P = 0.02) although with a non-significant 31% relative risk reduction (RRR). RRR for death alone was 89% (95% CI, 14 to 99; P-value not reported). Number needed to treat to prevent 1 death or 1 hospitalization or death composite appears to be closely competitive to other agents having EUA in people with COVID-19. However, cost-wise molnupiravir is comparatively cheaper compared to all other agents. CONCLUSION: Molnupiravir could be a useful agent in non-pregnant unvaccinated adults with COVID-19 who are at increased risk of severity including hospitalization. However, it is effective only when used within 5-days of onset of symptoms. A 5-days course seems to be safe without any obvious short-term side effects.


Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Cytidine/analogs & derivatives , Hydroxylamines/therapeutic use , SARS-CoV-2 , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/therapeutic use , Aged , Alanine/analogs & derivatives , Alanine/therapeutic use , Animals , COVID-19/mortality , COVID-19 Vaccines , Cytidine/adverse effects , Cytidine/therapeutic use , Double-Blind Method , Drug Approval , Drug Combinations , Female , Hospitalization , Humans , Hydroxylamines/adverse effects , Lactams/therapeutic use , Leucine/therapeutic use , Male , Middle Aged , Nitriles/therapeutic use , Proline/therapeutic use , Ritonavir/therapeutic use , Severity of Illness Index , Treatment Outcome
7.
Environ Mol Mutagen ; 63(1): 37-63, 2022 01.
Article En | MEDLINE | ID: mdl-35023215

This review considers antiviral nucleoside analog drugs, including ribavirin, favipiravir, and molnupiravir, which induce genome error catastrophe in SARS-CoV or SARS-CoV-2 via lethal mutagenesis as a mode of action. In vitro data indicate that molnupiravir may be 100 times more potent as an antiviral agent than ribavirin or favipiravir. Molnupiravir has recently demonstrated efficacy in a phase 3 clinical trial. Because of its anticipated global use, its relative potency, and the reported in vitro "host" cell mutagenicity of its active principle, ß-d-N4-hydroxycytidine, we have reviewed the development of molnupiravir and its genotoxicity safety evaluation, as well as the genotoxicity profiles of three congeners, that is, ribavirin, favipiravir, and 5-(2-chloroethyl)-2'-deoxyuridine. We consider the potential genetic risks of molnupiravir on the basis of all available information and focus on the need for additional human genotoxicity data and follow-up in patients treated with molnupiravir and similar drugs. Such human data are especially relevant for antiviral NAs that have the potential of permanently modifying the genomes of treated patients and/or causing human teratogenicity or embryotoxicity. We conclude that the results of preclinical genotoxicity studies and phase 1 human clinical safety, tolerability, and pharmacokinetics are critical components of drug safety assessments and sentinels of unanticipated adverse health effects. We provide our rationale for performing more thorough genotoxicity testing prior to and within phase 1 clinical trials, including human PIG-A and error corrected next generation sequencing (duplex sequencing) studies in DNA and mitochondrial DNA of patients treated with antiviral NAs that induce genome error catastrophe via lethal mutagenesis.


Antiviral Agents/adverse effects , COVID-19 Drug Treatment , Cytidine/analogs & derivatives , DNA Damage/drug effects , Hydroxylamines/adverse effects , Nucleosides/adverse effects , SARS-CoV-2/genetics , Amides/adverse effects , Amides/therapeutic use , Antiviral Agents/therapeutic use , Cytidine/adverse effects , Cytidine/therapeutic use , Deoxyuridine/adverse effects , Deoxyuridine/analogs & derivatives , Deoxyuridine/therapeutic use , Genome, Human/drug effects , Humans , Hydroxylamines/therapeutic use , Mutagenesis/drug effects , Nucleosides/therapeutic use , Pyrazines/adverse effects , Pyrazines/therapeutic use , Ribavirin/adverse effects , Ribavirin/therapeutic use , SARS-CoV-2/drug effects
8.
N Engl J Med ; 386(6): 509-520, 2022 02 10.
Article En | MEDLINE | ID: mdl-34914868

BACKGROUND: New treatments are needed to reduce the risk of progression of coronavirus disease 2019 (Covid-19). Molnupiravir is an oral, small-molecule antiviral prodrug that is active against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). METHODS: We conducted a phase 3, double-blind, randomized, placebo-controlled trial to evaluate the efficacy and safety of treatment with molnupiravir started within 5 days after the onset of signs or symptoms in nonhospitalized, unvaccinated adults with mild-to-moderate, laboratory-confirmed Covid-19 and at least one risk factor for severe Covid-19 illness. Participants in the trial were randomly assigned to receive 800 mg of molnupiravir or placebo twice daily for 5 days. The primary efficacy end point was the incidence hospitalization or death at day 29; the incidence of adverse events was the primary safety end point. A planned interim analysis was performed when 50% of 1550 participants (target enrollment) had been followed through day 29. RESULTS: A total of 1433 participants underwent randomization; 716 were assigned to receive molnupiravir and 717 to receive placebo. With the exception of an imbalance in sex, baseline characteristics were similar in the two groups. The superiority of molnupiravir was demonstrated at the interim analysis; the risk of hospitalization for any cause or death through day 29 was lower with molnupiravir (28 of 385 participants [7.3%]) than with placebo (53 of 377 [14.1%]) (difference, -6.8 percentage points; 95% confidence interval [CI], -11.3 to -2.4; P = 0.001). In the analysis of all participants who had undergone randomization, the percentage of participants who were hospitalized or died through day 29 was lower in the molnupiravir group than in the placebo group (6.8% [48 of 709] vs. 9.7% [68 of 699]; difference, -3.0 percentage points; 95% CI, -5.9 to -0.1). Results of subgroup analyses were largely consistent with these overall results; in some subgroups, such as patients with evidence of previous SARS-CoV-2 infection, those with low baseline viral load, and those with diabetes, the point estimate for the difference favored placebo. One death was reported in the molnupiravir group and 9 were reported in the placebo group through day 29. Adverse events were reported in 216 of 710 participants (30.4%) in the molnupiravir group and 231 of 701 (33.0%) in the placebo group. CONCLUSIONS: Early treatment with molnupiravir reduced the risk of hospitalization or death in at-risk, unvaccinated adults with Covid-19. (Funded by Merck Sharp and Dohme; MOVe-OUT ClinicalTrials.gov number, NCT04575597.).


Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Cytidine/analogs & derivatives , Hydroxylamines/therapeutic use , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Antiviral Agents/adverse effects , COVID-19/virology , Cytidine/adverse effects , Cytidine/therapeutic use , Double-Blind Method , Female , Humans , Hydroxylamines/adverse effects , Male , Middle Aged , SARS-CoV-2/isolation & purification , Treatment Outcome , Viral Load , Young Adult
11.
J Inherit Metab Dis ; 44(6): 1463-1480, 2021 11.
Article En | MEDLINE | ID: mdl-34418116

Niemann-Pick disease type C (NPC) is a rare, genetic, progressive neurodegenerative disorder with high unmet medical need. We investigated the safety and efficacy of arimoclomol, which amplifies the heat shock response to target NPC protein misfolding and improve lysosomal function, in patients with NPC. In a 12-month, prospective, randomised, double-blind, placebo-controlled, phase 2/3 trial (ClinicalTrials.gov identifier: NCT02612129), patients (2-18 years) were randomised 2:1 to arimoclomol:placebo, stratified by miglustat use. Routine clinical care was maintained. Arimoclomol was administered orally three times daily. The primary endpoint was change in 5-domain NPC Clinical Severity Scale (NPCCSS) score from baseline to 12 months. Fifty patients enrolled; 42 completed. At month 12, the mean progression from baseline in the 5-domain NPCCSS was 0.76 with arimoclomol vs 2.15 with placebo. A statistically significant treatment difference in favour of arimoclomol of -1.40 (95% confidence interval: -2.76, -0.03; P = .046) was observed, corresponding to a 65% reduction in annual disease progression. In the prespecified subgroup of patients receiving miglustat as routine care, arimoclomol resulted in stabilisation of disease severity over 12 months with a treatment difference of -2.06 in favour of arimoclomol (P = .006). Adverse events occurred in 30/34 patients (88.2%) receiving arimoclomol and 12/16 (75.0%) receiving placebo. Fewer patients had serious adverse events with arimoclomol (5/34, 14.7%) vs placebo (5/16, 31.3%). Treatment-related serious adverse events (n = 2) included urticaria and angioedema. Arimoclomol provided a significant and clinically meaningful treatment effect in NPC and was well tolerated.


Hydroxylamines/therapeutic use , Niemann-Pick Disease, Type C/drug therapy , Adolescent , Child , Child, Preschool , Disease Progression , Double-Blind Method , Female , Humans , Hydroxylamines/adverse effects , Internationality , Male , Niemann-Pick Disease, Type C/genetics , Prospective Studies , Severity of Illness Index , Treatment Outcome , Young Adult
12.
Invest New Drugs ; 39(1): 142-151, 2021 02.
Article En | MEDLINE | ID: mdl-32556884

Temozolomide (TMZ) generates DNA adducts that are repaired by direct DNA and base excision repair mechanisms. Methoxyamine (MX, TRC-102) potentiates TMZ activity by binding to apurinic and apyrimidinic (AP) sites after removal of N3-methyladenine and N7-methylguanine, inhibiting site recognition of AP endonuclease. We conducted a phase I trial to determine the maximum tolerated dose and dose-limiting toxicities (DLTs) of intravenous MX when given with oral TMZ. Patients with advanced solid tumors and progression on standard treatment were enrolled to a standard 3 + 3 dose escalation trial assessing escalating doses of TMZ and MX. Tumor response was assessed per RECIST and adverse events (AEs) by CTCAEv3. Pharmacokinetics (PK) of MX and COMET assays on peripheral blood mononuclear cells were performed. 38 patients were enrolled-median age 59.5 years (38-76), mean number of cycles 2.9 [1-13]. No DLTs were observed. Cycle 1 grade 3 AEs included fatigue, lymphopenia, anemia, INR, leukopenia, neutropenia, allergic reaction, constipation, psychosis and paranoia. Cycle 2-13 grade 4 AEs included thrombocytopenia and confusion. A partial response was seen in 1 patient with a pancreatic neuroendocrine tumor (PNET) and six additional patients, each with different tumor types, demonstrated prolonged stable disease. MX PK was linear with dose and was not affected by concomitant TMZ. TMZ 200 mg/m2 daily × 5 may be safely administered with MX 150 mg/m2 intravenously once on day 1 with minimal toxicity. Further studies assessing this drug combination in select tumor types where temozolomide has activity may be warranted.


Antineoplastic Agents, Alkylating/therapeutic use , Hydroxylamines/therapeutic use , Neoplasms/drug therapy , Temozolomide/therapeutic use , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Area Under Curve , DNA Repair/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Half-Life , Humans , Hydroxylamines/administration & dosage , Hydroxylamines/adverse effects , Hydroxylamines/pharmacokinetics , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Middle Aged , Temozolomide/adverse effects , Temozolomide/pharmacokinetics
13.
Neurology ; 90(7): e565-e574, 2018 02 13.
Article En | MEDLINE | ID: mdl-29367439

OBJECTIVE: To examine the safety and tolerability as well as the preliminary efficacy of arimoclomol, a heat shock protein co-inducer that promotes nascent protein folding, in patients with rapidly progressive SOD1 amyotrophic lateral sclerosis (ALS). METHODS: This was a double-blind, placebo-controlled trial in which patients with rapidly progressive SOD1-mutant ALS were randomized 1:1 to receive arimoclomol 200 mg tid or matching placebo for up to 12 months. Study procedures were performed using a mix of in-person and remote assessments. Primary outcome was safety and tolerability. Secondary outcome was efficacy, with survival as the principal measure. Additional efficacy measures were the rates of decline of the Revised ALS Functional Rating Scale (ALSFRS-R) and percent predicted forced expiratory volume in 6 seconds (FEV6), and the Combined Assessment of Function and Survival (CAFS). RESULTS: Thirty-eight participants were randomized. Thirty-six (19 placebo, 17 arimoclomol) were included in the prespecified intent-to-treat analysis. Apart from respiratory function, groups were generally well-balanced at baseline. Adverse events occurred infrequently, and were usually mild and deemed unlikely or not related to study drug. Adjusting for riluzole and baseline ALSFRS-R, survival favored arimoclomol with a hazard ratio of 0.77 (95% confidence interval [CI] 0.32-1.80). ALSFRS-R and FEV6 declined more slowly in the arimoclomol group, with treatment differences of 0.5 point/month (95% CI -0.63 to 1.63) and 1.24 percent predicted/month (95% CI -2.77 to 5.25), respectively, and the CAFS similarly favored arimoclomol. CONCLUSIONS: This study provides Class II evidence that arimoclomol is safe and well-tolerated at a dosage of 200 mg tid for up to 12 months. Although not powered for therapeutic effect, the consistency of results across the range of prespecified efficacy outcome measures suggests a possible therapeutic benefit of arimoclomol. CLINICALTRIALSGOV IDENTIFIER: NCT00706147. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that arimoclomol is safe and well-tolerated at a dosage of 200 mg tid for up to 12 months. The study lacked the precision to conclude, or to exclude, an important therapeutic benefit of arimoclomol.


Amyotrophic Lateral Sclerosis/drug therapy , Hydroxylamines/therapeutic use , Neuroprotective Agents/therapeutic use , Adult , Aged , Amyotrophic Lateral Sclerosis/genetics , Disease Progression , Double-Blind Method , Female , Humans , Hydroxylamines/adverse effects , Male , Middle Aged , Neuroprotective Agents/adverse effects , Severity of Illness Index , Superoxide Dismutase-1/genetics , Survival Analysis , Treatment Outcome
14.
Diabetes Obes Metab ; 19(7): 936-943, 2017 07.
Article En | MEDLINE | ID: mdl-28155245

AIM: To determine the effect of Scriptaid, a compound that can replicate aspects of the exercise adaptive response through disruption of the class IIa histone deacetylase (HDAC) corepressor complex, on muscle insulin action in obesity. MATERIALS AND METHODS: Diet-induced obese mice were administered Scriptaid (1 mg/kg) via daily intraperitoneal injection for 4 weeks. Whole-body and skeletal muscle metabolic phenotyping of mice was performed, in addition to echocardiography, to assess cardiac morphology and function. RESULTS: Scriptaid treatment had no effect on body weight or composition, but did increase energy expenditure, supported by increased lipid oxidation, while food intake was also increased. Scriptaid enhanced the expression of oxidative genes and proteins, increased fatty acid oxidation and reduced triglycerides and diacylglycerides in skeletal muscle. Furthermore, ex vivo insulin-stimulated glucose uptake by skeletal muscle was enhanced. Surprisingly, heart weight was reduced in Scriptaid-treated mice and was associated with enhanced expression of genes involved in oxidative metabolism in the heart. Scriptaid also improved indices of both diastolic and systolic cardiac function. CONCLUSION: These data show that pharmacological targeting of the class IIa HDAC corepressor complex with Scriptaid could be used to enhance muscle insulin action and cardiac function in obesity.


Cardiotonic Agents/therapeutic use , Energy Metabolism/drug effects , Heart/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Hydroxylamines/therapeutic use , Muscle, Skeletal/drug effects , Obesity/drug therapy , Quinolines/therapeutic use , Animals , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/therapeutic use , Cardiotonic Agents/adverse effects , Diet, High-Fat/adverse effects , Echocardiography , Echocardiography, Doppler , Gene Expression Profiling , Gene Expression Regulation/drug effects , Heart/diagnostic imaging , Heart/physiopathology , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/adverse effects , Hydroxylamines/adverse effects , Insulin Resistance , Male , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myocardium/pathology , Obesity/etiology , Obesity/pathology , Obesity/physiopathology , Organ Size , Quinolines/adverse effects
15.
Invest New Drugs ; 31(3): 714-23, 2013 Jun.
Article En | MEDLINE | ID: mdl-23054206

INTRODUCTION: TRC102 potentiates the activity of cancer therapies that induce base excision repair (BER) including antimetabolite and alkylating agents. TRC102 rapidly and covalently binds to apurinic/apyrimidinic (AP) sites generated during BER, and TRC102-bound DNA causes topoisomerase II-dependent irreversible strand breaks and apoptosis. This study assessed the safety, maximum-tolerated dose (MTD), pharmacokinetics and pharmacodynamics of TRC102 alone and in combination with pemetrexed. PURPOSE: Patients with advanced solid tumors received oral TRC102 daily for 4 days. Two weeks later, patients began standard-dose pemetrexed on day 1 in combination with oral TRC102 on days 1 to 4. The pemetrexed-TRC102 combination was repeated every 3 weeks until disease progression. METHODS: Twenty-eight patients were treated with TRC102 at 15, 30, 60 or 100 mg/m(2)/d. The MTD was exceeded at 100 mg/m(2)/d due to grade 3 anemia in 50 % of patients. TRC102 exposure increased in proportion to dose with a mean t1/2 of 28 h. A pharmacodynamic assay confirmed that TRC102 binds to pemetrexed-induced AP sites at all doses studied. Stable disease or better was achieved in 15 of 25 patients evaluable for response (60 %), including one patient with recurrent metastatic oropharyngeal carcinoma that expressed high levels of thymidylate synthase, who achieved a partial response and was progression free for 14 months. CONCLUSIONS: When administered with pemetrexed, the maximum tolerated dose of oral TRC102 is 60 mg/m(2)/d for 4 days. Randomized controlled studies are planned to evaluate the clinical benefit of adding TRC102 to pemetrexed and other agents that induce BER.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Anemia/chemically induced , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , DNA Repair , Female , Glutamates/administration & dosage , Glutamates/adverse effects , Guanine/administration & dosage , Guanine/adverse effects , Guanine/analogs & derivatives , Humans , Hydroxylamines/administration & dosage , Hydroxylamines/adverse effects , Hydroxylamines/pharmacokinetics , Male , Maximum Tolerated Dose , Middle Aged , Pemetrexed , Young Adult
17.
IDrugs ; 13(7): 482-96, 2010 Jul.
Article En | MEDLINE | ID: mdl-20582873

Recent years have seen an explosion of research into increasingly prevalent neurodegenerative diseases. Arimoclomol (BRX-220), being developed by CytRx Corp, is an oral therapeutic candidate for the treatment of amyotrophic lateral sclerosis (ALS), the most common form of motor neuron disease. ALS is a fatal, incurable disorder, which can present as sporadic (90 to 95% of cases) or familial (5 to 10% of cases) forms. The etiology of sporadic ALS remains unknown and much of the understanding of ALS pathogenesis has been derived through study of its familial forms; in particular, through study of autosomal dominant mutations in the SOD1 (copper/zinc superoxide dismutase) gene, which cause approximately 20% of familial ALS cases. Under conditions of excessive stress, arimoclomol induces amplification of the cytoprotective heat shock response in order to protect motor neurons from death. Comprehensive in vivo and in vitro studies demonstrated its effect in the prevention of neuronal loss and promotion of motor neuron survival, even after symptom onset. Clinical trials have reported good tolerability and safety. This paper discusses the rationale for arimoclomol use in ALS, the preclinical and clinical evidence collected to date, the likelihood of its promising preclinical results translating to humans, and the relevance of this research for neurodegeneration as a whole.


Amyotrophic Lateral Sclerosis/drug therapy , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Heat-Shock Proteins/metabolism , Hydroxylamines/pharmacology , Hydroxylamines/therapeutic use , Up-Regulation/drug effects , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Chaperonins/agonists , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacokinetics , Heat-Shock Response/drug effects , Humans , Hydroxylamines/adverse effects , Hydroxylamines/pharmacokinetics , Nerve Degeneration/drug therapy , Nerve Degeneration/prevention & control , Neurons/drug effects , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Stress, Physiological
18.
Expert Opin Investig Drugs ; 18(12): 1907-18, 2009 Dec.
Article En | MEDLINE | ID: mdl-19938902

Arimoclomol, an amplifier of heat shock protein expression involved in cellular stress response, has emerged as a potential therapeutic candidate in amyotrophic lateral sclerosis (ALS) in recent years. Treatment with arimoclomol was reported to improve survival and muscle function in a mouse model of motor neuron disease. Several single- and multiple-dose safety studies have been completed in healthy control subjects. A 3-month Phase IIa study in people with ALS demonstrated safety at dosages up to 300 mg/day and another study is currently recruiting participants with familial ALS caused by mutations in the superoxide dismutase gene. We review the rationale for testing arimoclomol in sporadic and familial ALS in the context of available safety and pharmacokinetic data. Published and unpublished literature relative to the drug in the past two decades is discussed. The current review attempts to bring together our existing understanding of the actions of arimoclomol with the disease profile of ALS. The pharmacological profile of arimoclomol and the available preclinical data make it a promising therapeutic possibility in ALS.


Amyotrophic Lateral Sclerosis/drug therapy , Hydroxylamines/pharmacology , Hydroxylamines/therapeutic use , Animals , Clinical Trials as Topic , Cytoprotection/drug effects , Drug Approval , Drug Evaluation, Preclinical , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/drug effects , Humans , Hydroxylamines/adverse effects , Hydroxylamines/pharmacokinetics
19.
Muscle Nerve ; 38(1): 837-44, 2008 Jul.
Article En | MEDLINE | ID: mdl-18551622

Arimoclomol is an investigational drug for amyotrophic lateral sclerosis (ALS) that amplifies heat shock protein gene expression during cell stress. The objectives of the present study were to assess the safety, tolerability, and pharmacokinetics of arimoclomol in ALS. Eighty-four participants with ALS received arimoclomol at one of three oral doses (25, 50, or 100 mg three times daily) or placebo. The primary outcome measure was safety and tolerability. A subset of 44 participants provided serum and cerebrospinal fluid (CSF) samples for pharmacokinetic analysis. Participants who completed 12 weeks of treatment could enroll in a 6-month open-label study. Arimoclomol at doses up to 300 mg/day was well tolerated and safe. Arimoclomol resulted in dose-linear pharmacologic exposures and the half-life did not change with continued treatment. Arimoclomol CSF levels increased with dose. Arimoclomol was shown to be safe, and it crosses the blood-brain barrier. Serum pharmacokinetic profiles support dosing of three times per day. An efficacy study in ALS is planned.


Amyotrophic Lateral Sclerosis/drug therapy , Hydroxylamines/administration & dosage , Hydroxylamines/adverse effects , Aged , Amyotrophic Lateral Sclerosis/metabolism , Blood-Brain Barrier , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Hydroxylamines/pharmacokinetics , Male , Middle Aged
20.
Toxicology ; 216(2-3): 129-39, 2005 Dec 15.
Article En | MEDLINE | ID: mdl-16129536

5-(Aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954) is an anti-tumour prodrug which recently entered clinical trials in combination with Escherichia coli nitroreductase in a gene-directed enzyme prodrug therapy (GDEPT) context. A Phase I trial of the prodrug, however, revealed dose-limiting hepatotoxicity (transaminitis). The aim of this study was to find out whether the prodrug undergoes reductive metabolism in human liver to cytotoxic metabolites which may contribute to this clinical toxicity. CB 1954 (2.5-250 microM) was incubated with human liver preparations (2-8 mg/mL of S9, cytosolic or microsomal proteins) in the presence of NAD(P)H (1 mM). The NADH- and NADPH-dependent formation of both 2- and 4-nitroreduction products was demonstrated, with NADPH being the preferred cofactor, by HPLC and mass spectrometry. The major metabolite formed in all three human liver preparations was the 4-hydroxylamine, a potent DNA cross-linking cytotoxin. The 2-hydroxylamine and 2-amine metabolites were also detected, both of which have also been demonstrated to be highly cytotoxic. 2-Nitroreduction was far greater in S9 compared with cytosol and was not detected in microsomal preparations. Although 2- and 4-nitroreduction of CB 1954 was inhibited under hyperoxic conditions, substantial metabolism was observed under atmospheric oxygen levels. These studies demonstrate that human liver is capable of aerobic reductive bioactivation of CB 1954 to cytotoxic metabolites in vitro, possibly involving multiple enzymes, which may account for the clinical hepatotoxicity observed.


Aziridines/metabolism , Liver/metabolism , Nitroreductases/metabolism , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/metabolism , Aziridines/antagonists & inhibitors , Carbon Monoxide/pharmacology , Chromatography, High Pressure Liquid , Clinical Trials, Phase I as Topic , Cytosol/enzymology , Cytotoxins/metabolism , Dicumarol/metabolism , Dicumarol/pharmacology , Escherichia coli/enzymology , Humans , Hydroxylamines/adverse effects , Hydroxylamines/chemistry , Hydroxylamines/metabolism , Hypoxia , Liver/drug effects , Mass Spectrometry , Microsomes, Liver/metabolism , NAD/drug effects , NAD/metabolism , NADP/drug effects , NADP/metabolism , Nitroreductases/antagonists & inhibitors , Prodrugs/metabolism
...