Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 48
1.
Pharmacol Res Perspect ; 12(3): e1200, 2024 Jun.
Article En | MEDLINE | ID: mdl-38655895

Cerebral cavernous malformation (CCM) has variable clinical symptoms, including potentially fatal hemorrhagic stroke. Treatment options are very limited, presenting a large unmet need. REC-994 (also known as tempol), identified as a potential treatment through an unbiased drug discovery platform, is hypothesized to treat CCMs through a reduction in superoxide, a reactive oxygen species. We investigated the safety, tolerability, and pharmacokinetic profile of REC-994 in healthy volunteers. Single- and multiple-ascending dose (SAD and MAD, respectively) studies were conducted in adult volunteers (ages 18-55). SAD study participants received an oral dose of REC-994 or placebo. MAD study participants were randomized 3:1 to oral doses of REC-994 or matching placebo, once daily for 10 days. Thirty-two healthy volunteers participated in the SAD study and 52 in the MAD study. Systemic exposure increased in proportion to REC-994 dose after single doses of 50-800 mg and after 10 days of dosing over the 16-fold dose range of 50-800 mg. Median Tmax and mean t1/2 were independent of dose in both studies, and the solution formulation was more rapidly absorbed. REC-994 was well tolerated. Treatment-emergent adverse effects across both studies were mild and transient and resolved by the end of the study. REC-994 has a favorable safety profile and was well tolerated in single and multiple doses up to 800 mg with no dose-limiting adverse effects identified. Data support conducting a phase 2 clinical trial in patients with symptomatic CCM.


Cyclic N-Oxides , Dose-Response Relationship, Drug , Spin Labels , Humans , Adult , Male , Female , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/pharmacokinetics , Cyclic N-Oxides/adverse effects , Young Adult , Middle Aged , Adolescent , Double-Blind Method , Healthy Volunteers , Oxidation-Reduction , Administration, Oral , Hemangioma, Cavernous, Central Nervous System/drug therapy
2.
Clin Pharmacol Ther ; 111(2): 391-403, 2022 02.
Article En | MEDLINE | ID: mdl-33998672

This study evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of BIA 10-2474, a fatty acid amide hydrolase (FAAH) inhibitor, after first administration to healthy male and female participants. Participants (n = 116) were recruited into this phase I, double-blind, randomized, placebo-controlled, single ascending dose and multiple ascending dose (10-day) study. The primary outcome was the safety and tolerability of BIA 10-2474. Secondary outcomes were pharmacokinetics of BIA 10-2474 and pharmacodynamics, considering plasma concentrations of anandamide and three other fatty acid amides (FAAs) and leukocyte FAAH activity. Single oral doses of 0.25-100 mg and repeated oral doses of 2.5-50 mg were evaluated. BIA 10-2474 was well tolerated up to 100 mg as a single dose and up to 20 mg once daily for 10 days. In the cohort receiving repeated administrations of 50 mg, there were central nervous system adverse events in five of six participants, one with fatal outcome, which led to early termination of the study. BIA 10-2474 showed a linear relationship between dose and area under plasma concentration-time curve (AUC) across the entire dose range and reached steady state within 5-6 days of administration, with an accumulation ratio, based on AUC0-24h , of <2 on Day 10. BIA 10-2474 was rapidly absorbed with a mean terminal elimination half-life of 8-10 hours (Day 10). BIA 10-2474 caused reversible, dose-related increases in plasma FAAs. In conclusion, we propose that these data, as well as the additional data generated since the clinical trial was stopped, do not provide a complete mechanistic explanation for the tragic fatality.


Amidohydrolases/antagonists & inhibitors , Central Nervous System/drug effects , Cyclic N-Oxides/adverse effects , Enzyme Inhibitors/adverse effects , Pyridines/adverse effects , Administration, Oral , Central Nervous System/physiopathology , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/pharmacokinetics , Double-Blind Method , Drug Administration Schedule , Drug Dosage Calculations , Early Termination of Clinical Trials , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , France , Healthy Volunteers , Humans , Male , Patient Safety , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Risk Assessment , Risk Factors
3.
Eur J Clin Pharmacol ; 76(9): 1237-1243, 2020 Sep.
Article En | MEDLINE | ID: mdl-32488334

AIM: The aim of this article is to understand the pros and cons of various methods involved in first-in-human (FIH) dose calculation and act decisively in dose escalations when calculating the maximum tolerated dose. SUBJECTS AND METHODS: We reviewed early phase clinical trials for methods of FIH dose and dose-escalation steps and discuss them in line with existing guidelines. We also reviewed the clinical trial registry to recognize trends in trial registration in recent years and after a massive failure in a few trials. RESULTS: Phase 1 trials of TGN 1412 and BIA10-2474 would always be remembered as catastrophes for pharmaceutical development plans. Quite often than not, healthy human volunteers are the guinea pigs in this stage of drug development. And, the most important aspect of designing an early phase study is deciding upon the dose to be started with, apart from the selection of cohort and escalation steps. The common principles used for FIH dose calculation include no observed adverse effect level, minimum anticipated biological effect level, pharmacologically active dose, pharmacokinetic/pharmacodynamic approach, and similar drug comparison approach. CONCLUSION: Early phase clinical trials are basically foundation stones on which lies the entire onus of the later stages of development. Deciding FIH dose is a crucial step that necessitates the incorporation of detailed data from the preclinical stages and application of the most conservative approach for the safety/benefit of the volunteers in these studies.


Clinical Trials, Phase I as Topic/methods , Maximum Tolerated Dose , Research Design , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Clinical Trials, Phase I as Topic/standards , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/adverse effects , Dose-Response Relationship, Drug , Humans , Pharmaceutical Preparations/administration & dosage , Pyridines/administration & dosage , Pyridines/adverse effects
4.
Mutat Res Rev Mutat Res ; 782: 108283, 2019.
Article En | MEDLINE | ID: mdl-31843137

Immuno-spin trapping detection of DNA radicals with the nitrone spin trap 5,5-dimethyl-1-pyrrloine N-oxide (DMPO) has made important contributions towards the understanding of DNA radicalization and genotoxicity at sites of inflammation. At sites of inflammation, one-electron oxidants and chloramines decay induce oxidation of genomic DNA, genotoxicity and cell transformation. Radicalization of DNA can result in either single- or double-strand breaks, or end-oxidation products at the sugar or bases. If not repaired, these modifications can lead to mutations and cell transformation. If trapped with DMPO, DNA-centered radical decay and subsequent formation of end-oxidation products are blocked. Herein we discuss recent literature regarding the use of immuno-spin trapping with DMPO to study DNA-centered radicals and their involvement in genotoxicity. This technique has shown the critical role of DNA radicalization in 8-oxo-dG formation and DNA strand breaks in isolated DNA, cells and in whole animals. Combination of technologies, including immuno-spin trapping and powerful chromatographic and sequencing techniques are needed to move forward the field towards the detection of specific genes that are susceptible to oxidative damage in cells located at sites of inflammation. This is important in order to provide novel information about genotoxicity mechanisms, as well as therapeutic possibilities of DMPO or its derivatives for preventing DNA-centered radical-mediated carcinogenesis.


Cyclic N-Oxides/adverse effects , DNA Damage/drug effects , DNA/drug effects , Free Radicals/chemistry , Mutagens/adverse effects , Nitrogen Oxides/adverse effects , Nitrogen Oxides/chemistry , Animals , Inflammation/genetics , Spin Trapping/methods
5.
Environ Mol Mutagen ; 60(7): 588-593, 2019 08.
Article En | MEDLINE | ID: mdl-31001845

2-Hydroxypyridine N-oxide (HOPO) is an important coupling reagent used in pharmaceutical synthesis. Our laboratory previously reported HOPO as equivocal in the Ames assay following extensive testing of multiple lots of material. Given the lack of reproducibility between lots of material and the weak increase in revertants observed, it was concluded that it would be highly unlikely that HOPO would pose a mutagenic risk in vivo. The purpose of the current investigation was to assess experimentally in rats the mutagenic (Pig-a mutation induction) and more broadly genotoxic (micronucleus and comet induction) potential of HOPO. Rats were administered HOPO (0, 50, 150, 300, and 500 mg/kg/day) by oral gavage for 28 days. At the end of study, the following parameters were assessed: frequency of Pig-a mutant red blood cells and reticulocytes, frequency of peripheral blood micronuclei, and the incidence of comet formation in liver. Toxicokinetic data collected on study Days 1 and 28 demonstrated systemic exposure to HOPO. Although there were no overt clinical signs, animals treated with HOPO showed a dose-related decrease in body weight gain. There were no increases observed in any of the genotoxicity endpoints assessed. The results from this study further support the conclusion that in the context of pharmaceutical synthesis, HOPO should not be considered a mutagenic impurity but rather controlled as a normal process-related impurity. Environ. Mol. Mutagen. 2019. © 2019 Wiley Periodicals, Inc.


Cyclic N-Oxides/adverse effects , Mutagenesis/drug effects , Mutagens/adverse effects , Pyridines/adverse effects , Animals , Erythrocytes/drug effects , Liver/drug effects , Male , Mutagenicity Tests/methods , Mutation/drug effects , Rats , Rats, Wistar , Reproducibility of Results , Reticulocytes/drug effects
6.
ChemMedChem ; 13(20): 2177-2188, 2018 10 22.
Article En | MEDLINE | ID: mdl-30113139

Fatty acid amide hydrolase (FAAH) can be targeted for the treatment of pain associated with various medical conditions. Herein we report the design and synthesis of a novel series of heterocyclic-N-carboxamide FAAH inhibitors that have a good alignment of potency, metabolic stability and selectivity for FAAH over monoacylglycerol lipase (MAGL) and carboxylesterases (CEs). Lead optimization efforts carried out with benzotriazolyl- and imidazolyl-N-carboxamide series led to the discovery of clinical candidate 8 l (3-(1-(cyclohexyl(methyl)carbamoyl)-1H-imidazol-4-yl)pyridine 1-oxide; BIA 10-2474) as a potent and long-acting inhibitor of FAAH. However, during a Phase I clinical trial with compound 8 l, unexpected and unpredictable serious neurological adverse events occurred, affecting five healthy volunteers, including the death of one subject.


Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Cyclic N-Oxides/pharmacology , Enzyme Inhibitors/pharmacology , Pyridines/pharmacology , Administration, Oral , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/chemistry , Animals , Brain/drug effects , Brain/metabolism , Clinical Trials, Phase I as Topic , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/chemistry , Drug Design , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/chemistry , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/chemistry , Rats , Structure-Activity Relationship
7.
Circ Cardiovasc Interv ; 11(6): e006084, 2018 06.
Article En | MEDLINE | ID: mdl-29895599

BACKGROUND: Monitoring anticoagulation with activated clotting time (ACT) has been proposed to reduce ischemic or bleeding events. However, the value of using ACT to improve outcomes is uncertain. This study sought to determine the relationship between ACT and outcomes during percutaneous coronary intervention in patients with non-ST-segment-elevation acute coronary syndrome (NSTE-ACS) treated by unfractionated heparin with GPIs (glycoprotein IIb/IIIa inhibitors). METHODS AND RESULTS: From the randomized TAO trial (Treatment of Acute Coronary Syndromes With Otamixaban), we analyzed the value of ACT to predict ischemic and bleeding outcomes in the 3275 patients receiving unfractionated heparin plus eptifibatide. Ischemic and safety outcomes were analyzed according to ACT to determine the best threshold. Median peak ACT was 225 s. There was no correlation (r=-0.02; P=0.24) between the unfractionated heparin dose received and the ACT value before percutaneous coronary intervention. There was no evidence of a nonlinear association between ACT and either ischemic or bleeding events (P=0.66; P=0.07). No threshold was found to predict ischemic complications. Conversely, increased bleeding was observed with ACT >230 s with an optimal threshold of ACTs ≥250 s (4.53% versus 6.17%; odds ratio, 1.46; 95% confidence interval, 1.04-2.06; P=0.028). This optimal threshold varied according to access site: ≥250 s (6.86% versus 10.18%; odds ratio, 1.57; 95% confidence interval, 1.00-2.45; P=0.047) by femoral approach and ≥290 s (2.86% versus 5.43%; odds ratio, 2.24; 95% confidence interval, 1.05-4.44; P=0.027) by radial approach. CONCLUSIONS: In the TAO trial, peak procedural ACT ≥250 s was associated with increased bleeding risk in non-ST-segment-elevation acute coronary syndrome patients treated with unfractionated heparin plus GPIs. This threshold was increased to 290 s when performing radial approach. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov. Unique identifier: NCT01076764.


Acute Coronary Syndrome/therapy , Anticoagulants/adverse effects , Blood Coagulation/drug effects , Drug Monitoring/methods , Eptifibatide/adverse effects , Hemorrhage/chemically induced , Heparin/adverse effects , Non-ST Elevated Myocardial Infarction/therapy , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/adverse effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Whole Blood Coagulation Time , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnosis , Aged , Anticoagulants/administration & dosage , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/adverse effects , Drug Dosage Calculations , Eptifibatide/administration & dosage , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/adverse effects , Female , Heparin/administration & dosage , Humans , Male , Middle Aged , Non-ST Elevated Myocardial Infarction/blood , Non-ST Elevated Myocardial Infarction/diagnosis , Percutaneous Coronary Intervention/adverse effects , Platelet Aggregation Inhibitors/administration & dosage , Predictive Value of Tests , Pyridines/administration & dosage , Pyridines/adverse effects , Risk Factors , Time Factors , Treatment Outcome
8.
Rev Recent Clin Trials ; 13(4): 252-256, 2018.
Article En | MEDLINE | ID: mdl-29779485

BACKGROUND: Globally, there have been tremendous efforts by regulatory authorities to make clinical trials safer by making stringent clinical trial regulations. Despite this, we witnessed several tragic events. TGN1412 and BIA 10-2474 phase I trials are infamous trails in which healthy volunteers either succumbed to severe adverse effects or faced irreversible impairments of the test drug. Such afflictions in clinical trials are not only turbulent to the image of pharmaceutical industry but it also conveys dispiriting message for clinical trial participants. OBJECTIVE: To make clinical trials safer for participants, some regulatory changes are warranted. METHODS: Some stipulated measures to improve safety of clinical trial participants include inclusion of patients instead of healthy volunteers in phase I clinical trials, all compounds which are used for first time in humans should be considered as high risk compounds, amendments in first in human clinical trial design to N of 1 randomized control trial in place of 6+2 design with the sequential dosing of subjects both within and between cohorts and the individual patient pharmacokinetic and pharmacodynamic data should be used to calculate sequential dosing. Besides these, there should be appropriate process for systematic risk assessment involving the use of statistical techniques to select pertinent risk factors with high predictive values of studies or sites that may be procumbent to non-compliance. CONCLUSION: Inclusion of above mentioned measures in clinical trials are bound to make them safer and may help in pacifying the insecurity that has emerged among humans to participate in clinical trials.


Antibodies, Monoclonal, Humanized/adverse effects , Clinical Trials as Topic , Cyclic N-Oxides/adverse effects , Patient Safety , Pyridines/adverse effects , Research Design , Humans
9.
Br J Clin Pharmacol ; 83(11): 2339-2342, 2017 Nov.
Article En | MEDLINE | ID: mdl-28681444

This is a joint statement from individual pharmacology and pharmaceutical professionals acting in their own capacity, including members of the Alliance for Clinical Research Excellence and Safety (ACRES) and the International Society of Pharmacovigilance (ISoP). By building on the extensive pharmacological and regulatory investigations that already take place, we are calling for a fuller and more robust systems-based approach to the independent investigation of clinical research when serious incidents of harm occur, starting with first-in-human clinical trials. To complement existing activities and regulations, we propose an additional approach blending evidence derived from both pharmacological and organizational science, which addresses human factors and transparency, to enhance organizational learning and continuous improvement. As happens with investigations in other sectors of society, such as the chemical and aviation sector, this systems approach should be seen as an additional way to understand how problems occur and how they might be prevented in the future. We believe that repetition of potentially preventable and adverse outcomes during clinical research, by failing to identify and act upon all systematic vulnerabilities, is a situation that needs urgent change. As we will discuss further on, approaches based on applying systems theory and human factors are much more likely to improve objectivity and transparency, leading to better system design.


Delivery of Health Care/organization & administration , Human Experimentation , Pharmacovigilance , Quality Improvement/organization & administration , Systems Theory , Antibodies, Monoclonal, Humanized/adverse effects , Clinical Trials, Phase I as Topic , Cyclic N-Oxides/adverse effects , Delivery of Health Care/legislation & jurisprudence , Humans , Pyridines/adverse effects
10.
Science ; 356(6342): 1084-1087, 2017 06 09.
Article En | MEDLINE | ID: mdl-28596366

A recent phase 1 trial of the fatty acid amide hydrolase (FAAH) inhibitor BIA 10-2474 led to the death of one volunteer and produced mild-to-severe neurological symptoms in four others. Although the cause of the clinical neurotoxicity is unknown, it has been postulated, given the clinical safety profile of other tested FAAH inhibitors, that off-target activities of BIA 10-2474 may have played a role. Here we use activity-based proteomic methods to determine the protein interaction landscape of BIA 10-2474 in human cells and tissues. This analysis revealed that the drug inhibits several lipases that are not targeted by PF04457845, a highly selective and clinically tested FAAH inhibitor. BIA 10-2474, but not PF04457845, produced substantial alterations in lipid networks in human cortical neurons, suggesting that promiscuous lipase inhibitors have the potential to cause metabolic dysregulation in the nervous system.


Amidohydrolases/antagonists & inhibitors , Analgesics/pharmacology , Anti-Anxiety Agents/pharmacology , Cyclic N-Oxides/pharmacology , Neurons/drug effects , Pyridines/pharmacology , Analgesics/adverse effects , Analgesics/chemistry , Analgesics/metabolism , Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/metabolism , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Cross Reactions , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/metabolism , Humans , Neurons/metabolism , Protein Interaction Maps , Pyridazines/pharmacology , Pyridazines/therapeutic use , Pyridines/adverse effects , Pyridines/chemistry , Pyridines/metabolism , Urea/analogs & derivatives , Urea/pharmacology , Urea/therapeutic use
11.
J Clin Pharmacol ; 57(6): 690-703, 2017 06.
Article En | MEDLINE | ID: mdl-28387940

BIA 10-2474 (a fatty acid amide hydrolase inhibitor) was evaluated in a first-in-human phase 1 study in normal volunteers to assess safety/tolerability, pharmacokinetics, pharmacodynamics, and food effect. The dose-escalation process consisted of a single-ascending-dose phase (SAD) and multiple-ascending-dose phase (MAD). Prospective determination of the starting dose and maximal escalated dose was consistent with the usual clinical pharmacology principles for extrapolation of preclinical toxicology data to human equivalent doses. After only 5-6 days of multiple-dose administration of 50 mg daily in the MAD phase, several subjects became quite ill with central nervous system symptoms. One subject progressed to brain death within several days of symptom onset. Magnetic resonance imaging scans demonstrated signal abnormalities consistent with microbleeds affecting the hippocampus and pons, suggestive of possible cytotoxic or vasogenic edema compatible with a toxic/metabolic process. There were no findings at lower MAD doses or during the SAD phase. The toxicology program carried out in 4 preclinical species (mouse, rat, dog, and monkey) did not demonstrate significant neurotoxicity. The probable mechanism of neurologic toxicity demonstrated in humans at the 50-mg daily dose was inhibition of off-target cerebral receptors or through another mechanism. Additional recommendations have been proposed for future first-in-human studies to maximize subject safety. However, one must also accept the basic premise that, in general, first-in-human phase 1 studies are remarkably safe, and these rare events are not 100% avoidable during the drug development process.


Clinical Trials, Phase I as Topic , Amidohydrolases/antagonists & inhibitors , Animals , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/pharmacokinetics , Cyclic N-Oxides/pharmacology , Humans , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyridines/pharmacology
12.
Pharm Stat ; 16(2): 100-106, 2017 03.
Article En | MEDLINE | ID: mdl-28206702

By setting the regulatory-approved protocol for a suite of first-in-human studies on BIA 10-2474 against the subsequent French investigations, we highlight 6 key design and statistical issues, which reinforce recommendations by a Royal Statistical Society Working Party, which were made in the aftermath of cytokine release storm in 6 healthy volunteers in the United Kingdom in 2006. The 6 issues are dose determination, availability of pharmacokinetic results, dosing interval, stopping rules, appraisal by safety committee, and clear algorithm required if combining approvals for single and multiple ascending dose studies.


Cyclic N-Oxides/administration & dosage , Data Interpretation, Statistical , Pyridines/administration & dosage , Research Design , Algorithms , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/pharmacokinetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Drug and Narcotic Control , France , Humans , Pyridines/adverse effects , Pyridines/pharmacokinetics , United Kingdom
13.
J Physiol Biochem ; 73(2): 207-214, 2017 May.
Article En | MEDLINE | ID: mdl-27933463

Vasoconstriction within the renal medulla contributes to the development of hypertension. This study investigated the role of reactive oxygen species (ROS) in regulating renal medullary and cortical blood perfusion (MBP and CBP respectively) in both stroke-prone spontaneously hypertensive rats (SHRSP) and Wistar rats. CBP and MBP were measured using a laser-Doppler flow meter before and after intra-renal infusion of tempol, the superoxide dismutase (SOD) mimetic or tempol plus catalase, the hydrogen peroxide-degrading enzyme. Tempol infusion significantly elevated blood perfusion within the renal medulla (MBP) in both SHRSP (by 43 ± 7%, P < 0.001) and Wistar rats (by 17 ± 2%, P < 0.05) but the magnitude of the increase was significantly greater in the SHRSP (P < 0.01). When the enzyme catalase and tempol were co-infused, MBP was again significantly increased in SHRSP (by 57 ± 6%, P < 0.001) and Wistar rats (by 33 ± 6%, P < 0.001), with a significantly greater increase in perfusion being induced in the SHRSP relative to the Wistar rats (P < 0.01). Notably, this increase was significantly greater than in those animals infused with tempol alone (P < 0.01). These results suggest that ROS plays a proportionally greater role in reducing renal vascular compliance, particularly within the renal medulla, in normotensive and hypertensive animals, with effects being greater in the hypertensive animals. This supports the hypothesis that SHRSP renal vasculature might be subjected to elevated level of oxidative stress relative to normotensive animals.


Antihypertensive Agents/therapeutic use , Antioxidants/therapeutic use , Catalase/therapeutic use , Cyclic N-Oxides/therapeutic use , Hypertension/drug therapy , Kidney/drug effects , Renal Circulation/drug effects , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/adverse effects , Antioxidants/administration & dosage , Antioxidants/adverse effects , Catalase/administration & dosage , Catalase/adverse effects , Cattle , Cyclic N-Oxides/administration & dosage , Cyclic N-Oxides/adverse effects , Drug Therapy, Combination/adverse effects , Hypertension/metabolism , Hypertension/physiopathology , Hypodermoclysis , Kidney/blood supply , Kidney/metabolism , Kidney/physiopathology , Kidney Medulla/blood supply , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Kidney Medulla/physiopathology , Male , Oxidative Stress/drug effects , Rats, Inbred SHR , Rats, Wistar , Reactive Oxygen Species/metabolism , Spin Labels , Stroke/etiology , Stroke/prevention & control , Vascular Resistance/drug effects
14.
Biochem Biophys Res Commun ; 483(1): 502-508, 2017 01 29.
Article En | MEDLINE | ID: mdl-28007597

The investigational compound BIA 10-2474, designed as a long-acting and reversible inhibitor of fatty acid amide hydrolase for the treatment of neuropathic pain, led to the death of one participant and hospitalization of five others due to intracranial hemorrhage in a Phase I clinical trial. Putative off-target activities of BIA 10-2474 have been suggested to be major contributing factors to the observed neurotoxicity in humans, motivating our study's proteome-wide screening approach to investigate its polypharmacology. Accordingly, we performed an in silico screen against 80,923 protein structures reported in the Protein Data Bank. The resulting list of 284 unique human interactors was further refined using target-disease association analyses to a subset of proteins previously linked to neurological, intracranial, inflammatory, hemorrhagic or clotting processes and/or diseases. Eleven proteins were identified as potential targets of BIA 10-2474, and the two highest-scoring proteins, Factor VII and thrombin, both essential blood-clotting factors, were predicted to be inhibited by BIA 10-2474 and suggest a plausible mechanism of toxicity. Once this small molecule becomes commercially available, future studies will be conducted to evaluate the predicted inhibitory effect of BIA 10-2474 on blood clot formation specifically in the brain.


Analgesics/adverse effects , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/chemistry , Neurotoxicity Syndromes/metabolism , Proteome/metabolism , Pyridines/adverse effects , Pyridines/chemistry , Amidohydrolases/metabolism , Analgesics/chemistry , Analgesics/pharmacokinetics , Computational Biology/methods , Cyclic N-Oxides/pharmacokinetics , Humans , Molecular Docking Simulation , Proteome/chemistry , Pyridines/pharmacokinetics
17.
Appl Physiol Nutr Metab ; 42(1): 59-67, 2017 Jan.
Article En | MEDLINE | ID: mdl-28006433

Hypothalamic inflammation and increased oxidative stress are believed to be mechanisms that contribute to obesity. 4-Hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (tempol), a free radical scavenger, has been shown to reduce inflammation and oxidative stress. We hypothesized that brain infusion of tempol would reduce oxidative stress, and thus would reduce food intake and body weight and improve body composition in rats with age-related obesity and known elevated oxidative stress. Furthermore, we predicted an associated increase in markers of leptin signalling, including the silent mating type information regulator 2 homolog 1 (SIRT1)/5'AMP-activated protein kinase (AMPK) pathway and the signal transducer and activator of transcription 3 (STAT3) pathway. For this purpose, osmotic minipumps were placed in the intracerebroventricular region of young (3 months) and aged (23 months) male Fischer 344 x Brown Norway rats for the continuous infusion of tempol or vehicle for 2 weeks. Tempol significantly decreased (p < 0.01) nicotinamide adenine dinucleotide phosphate oxidase activity in the hypothalamus but failed to reduce food intake or weight gain and did not alter body composition. SIRT1 activity and Acetyl p53 were decreased and phosphorylation of AMPK was increased with age, but they were unchanged with tempol. Basal phosphorylation of STAT3 was unchanged with age or tempol. These results indicate that tempol decreases oxidative stress but fails to alter feeding behaviour, body weight, or body composition. Moreover, tempol does not modulate the SIRT1/AMPK/p53 pathway and does not change leptin signalling. Thus, a reduction in hypothalamic oxidative stress is not sufficient to reverse age-related obesity.


Aging , Cognitive Dysfunction/prevention & control , Cyclic N-Oxides/administration & dosage , Free Radical Scavengers/administration & dosage , Hypothalamus/drug effects , Nootropic Agents/administration & dosage , Oxidative Stress/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Cognitive Dysfunction/etiology , Crosses, Genetic , Cyclic N-Oxides/adverse effects , Cyclic N-Oxides/therapeutic use , Energy Intake/drug effects , Free Radical Scavengers/adverse effects , Free Radical Scavengers/therapeutic use , Hypothalamus/metabolism , Infusion Pumps, Implantable , Infusions, Intraventricular , Male , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Nootropic Agents/adverse effects , Nootropic Agents/therapeutic use , Obesity/drug therapy , Obesity/physiopathology , Rats, Inbred BN , Rats, Inbred F344 , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Spin Labels
18.
N Engl J Med ; 375(18): 1717-1725, 2016 11 03.
Article En | MEDLINE | ID: mdl-27806235

BACKGROUND: A decrease in fatty acid amide hydrolase (FAAH) activity increases the levels of endogenous analogues of cannabinoids, or endocannabinoids. FAAH inhibitors have shown analgesic and antiinflammatory activity in animal models, and some have been tested in phase 1 and 2 studies. In a phase 1 study, BIA 10-2474, an orally administered reversible FAAH inhibitor, was given to healthy volunteers to assess safety. METHODS: Single doses (0.25 to 100 mg) and repeated oral doses (2.5 to 20 mg for 10 days) of BIA 10-2474 had been administered to 84 healthy volunteers in sequential cohorts; no severe adverse events had been reported. Another cohort of participants was then assigned to placebo (2 participants) or 50 mg of BIA 10-2474 per day (6 participants). This report focuses on neurologic adverse events in participants in this final cohort. A total of 4 of the 6 participants who received active treatment consented to have their clinical and radiologic data included in this report. RESULTS: An acute and rapidly progressive neurologic syndrome developed in three of the four participants starting on the fifth day of drug administration. The main clinical features were headache, a cerebellar syndrome, memory impairment, and altered consciousness. Magnetic resonance imaging showed bilateral and symmetric cerebral lesions, including microhemorrhages and hyperintensities on fluid-attenuated inversion recovery and diffusion-weighted imaging sequences predominantly involving the pons and hippocampi. One patient became brain dead; the condition of two patients subsequently improved, but one patient had residual memory impairment, and the other patient had a residual cerebellar syndrome. One patient remained asymptomatic. CONCLUSIONS: An unanticipated severe neurologic disorder occurred after ingestion of BIA 10-2474 at the highest dose level used in a phase 1 trial. The underlying mechanism of this toxic cerebral syndrome remains unknown.


Amidohydrolases/antagonists & inhibitors , Cerebellar Diseases/chemically induced , Consciousness Disorders/chemically induced , Cyclic N-Oxides/adverse effects , Hippocampus/pathology , Memory Disorders/chemically induced , Pons/pathology , Pyridines/adverse effects , Acute Disease , Administration, Oral , Adult , Brain Death , Cerebellum/pathology , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/diagnosis , Cyclic N-Oxides/administration & dosage , Double-Blind Method , Gait Ataxia/chemically induced , Headache/chemically induced , Healthy Volunteers , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Pyridines/administration & dosage
...