Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.420
Filter
1.
Pathol Res Pract ; 257: 155290, 2024 May.
Article in English | MEDLINE | ID: mdl-38640781

ABSTRACT

The intricate relationship between smoking and the effects of the antiplatelet drug clopidogrel has been termed the "smoker's paradox". This paradox details the enhanced efficacy of clopidogrel in smokers compared to non-smokers. This review begins with an exploration of the proposed mechanisms of the smoker's paradox, particularly drawing attention to the induction of cytochrome P450 (CYP) isoenzymes via tobacco smoke, specifically the enzymes CYP1A2 and CYP2C19. Moreover, an investigation of the effects of genetic variability on the smoker's paradox was undertaken from both clinical and molecular perspectives, delving into the effects of ethnicity and genetic polymorphisms. The intriguing role of CYP1A2 genotypes and the response to clopidogrel in smoking and non-smoking populations was examined conferring insight into the individuality rather than universality of the smoker's paradox. CYP1A2 induction is hypothesised to elucidate the potency of smoking in exerting a counteracting effect in those taking clopidogrel who possess CYP2C19 loss of function polymorphisms. Furthermore, we assess the comparative efficacies of clopidogrel and other antiplatelet agents, namely prasugrel and ticagrelor. Studies indicated that prasugrel and ticagrelor provided a more consistent effect and further reduced platelet reactivity compared to clopidogrel within both smoking and non-smoking populations. Personalised dosing was another focus of the review considering patient comorbidities, genetic makeup, and smoking status with the objective of improving the antiplatelet response of those taking clopidogrel. In summation, this review provides insight into multiple areas of research concerning clopidogrel and the smoker's paradox taking into account proposed mechanisms, genetics, other antiplatelet agents, and personalised dosing.


Subject(s)
Clopidogrel , Platelet Aggregation Inhibitors , Smoking , Humans , Clopidogrel/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/pharmacology , Smoking/adverse effects , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19/metabolism , Smokers , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism
2.
Eur J Drug Metab Pharmacokinet ; 49(3): 367-381, 2024 May.
Article in English | MEDLINE | ID: mdl-38554232

ABSTRACT

BACKGROUND AND OBJECTIVE: Abrocitinib is an oral small-molecule Janus kinase (JAK)-1 inhibitor approved for the treatment of moderate-to-severe atopic dermatitis. In vitro studies indicated that abrocitinib is a weak time-dependent inhibitor of cytochrome P450 (CYP) 2C19/3A and a weak inducer of CYP1A2/2B6/2C19/3A. To assess the potential effect of abrocitinib on concomitant medications, drug-drug interaction (DDI) studies were conducted for abrocitinib with sensitive probe substrates of these CYP enzymes. The impact of abrocitinib on hormonal oral contraceptives (ethinyl estradiol and levonorgestrel), as substrates of CYP3A and important concomitant medications for female patients, was also evaluated. METHODS: Three Phase 1 DDI studies were performed to assess the impact of abrocitinib 200 mg once daily (QD) on the probe substrates of: (1) 1A2 (caffeine), 2B6 (efavirenz) and 2C19 (omeprazole) in a cocktail study; (2) 3A (midazolam); and (3) 3A (oral contraceptives). RESULTS: After multiple doses of abrocitinib 200 mg QD, there is a lack of effect on the pharmacokinetics of midazolam, efavirenz and contraceptives. Abrocitinib increased the area under the concentration time curve from 0 to infinity (AUCinf) and the maximum concentration (Cmax) of omeprazole by approximately 189 and 134%, respectively. Abrocitinib increased the AUCinf of caffeine by 40% with lack of effect on Cmax. CONCLUSIONS: Based on the study results, abrocitinib is a moderate inhibitor of CYP2C19. Caution should be exercised when using abrocitinib concomitantly with narrow therapeutic index medicines that are primarily metabolized by CYP2C19 enzyme. Abrocitinib is a mild inhibitor of CYP1A2; however, the impact is not clinically relevant, and no general dose adjustment is recommended for CYP1A2 substrates. Abrocitinib does not inhibit CYP3A or induce CYP1A2/2B6/2C19/3A and does not affect the pharmacokinetics of contraceptives. CLINICAL TRIALS REGISTRATION: ClinicalTrials.gov registration IDs: NCT03647670, NCT05067439, NCT03662516.


Subject(s)
Drug Interactions , Pyrimidines , Sulfonamides , Humans , Female , Adult , Young Adult , Pyrimidines/pharmacokinetics , Pyrimidines/administration & dosage , Cytochrome P-450 CYP1A2/metabolism , Male , Ethinyl Estradiol/pharmacokinetics , Healthy Volunteers , Contraceptives, Oral, Hormonal/pharmacokinetics , Cytochrome P-450 CYP2C19/metabolism , Levonorgestrel/pharmacokinetics , Levonorgestrel/administration & dosage , Contraceptives, Oral, Combined/pharmacokinetics , Contraceptives, Oral, Combined/administration & dosage , Middle Aged , Area Under Curve , Drug Combinations
3.
Environ Pollut ; 345: 123514, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38346634

ABSTRACT

N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine-quinone (6PPD-Q) is an ozonation product of the rubber antioxidant N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD). 6PPD-Q has recently been detected in various environmental media, which may enter the human body via inhalation and skin contact pathways. However, the human metabolism of 6PPD-Q has remained unknown. This study investigated the in vitro Cytochrome P450-mediated metabolism of 6PPD-Q in human and rat liver microsomes (HLMs and RLMs). 6PPD-Q was significantly metabolized at lower concentrations but slowed at high concentrations. The intrinsic clearance (CLint) of 6PPD-Q was 21.10 and 18.58 µL min-1 mg-1 protein of HLMs and RLMs, respectively, suggesting low metabolic ability compared with other reported pollutants. Seven metabolites and one intermediate were identified, and metabolites were predicted immunotoxic or mutagenic toxicity. Mono- and di-oxygenation reactions were the main phase I in vitro metabolic pathways. Enzyme inhibition experiments and molecular docking techniques were further used to reveal the metabolic mechanism. CYP1A2, 3A4, and 2C19, especially CYP1A2, play critical roles in 6PPD-Q metabolism in HLMs, whereas 6PPD-Q is extensively metabolized in RLMs. Our study is the first to demonstrate the in vitro metabolic profile of 6PPD-Q in HLMs and RLMs. The results will significantly contribute to future human health management targeting the emerging pollutant 6PPD-Q.


Subject(s)
Cytochrome P-450 CYP1A2 , Microsomes, Liver , Phenylenediamines , Humans , Rats , Animals , Cytochrome P-450 CYP1A2/metabolism , Microsomes, Liver/metabolism , Molecular Docking Simulation , Cytochrome P-450 Enzyme System/metabolism , Quinones , Kinetics
4.
Toxicol In Vitro ; 96: 105789, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38341109

ABSTRACT

Zearalenone (ZEN) is a mycoestrogen produced by Fusarium fungi. ZEN is a frequent contaminant in cereal-based products, representing significant health threat. The major reduced metabolites of ZEN are α-zearalenol (α-ZEL) and ß-zearalenol (ß-ZEL). Since the toxicokinetic interactions of ZEN/ZELs with cytochrome P450 enzymes (CYPs) and organic anion transporting polypeptides (OATPs) have been barely characterized, we examined these interactions applying in vitro models. ZEN and ZELs were relatively strong inhibitors of CYP3A4 and moderate inhibitors of CYP1A2 and CYP2C9. Both CYP1A2 and CYP3A4 decreased ZEN and ß-ZEL concentrations in depletion assays, while only CYP1A2 reduced α-ZEL levels. OATPs tested were strongly or moderately inhibited by ZEN and ZELs; however, these mycotoxins did not show higher cytotoxicity in OATP-overexpressing cells. Our results help the deeper understanding of the toxicokinetic/pharmacokinetic interactions of ZEN, α-ZEL, and ß-ZEL.


Subject(s)
Mycotoxins , Organic Anion Transporters , Zearalenone , Zeranol/analogs & derivatives , Zearalenone/toxicity , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System , Peptides
5.
Xenobiotica ; 54(2): 57-63, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38166553

ABSTRACT

This study assessed the effect of tubeimoside I on CYP1A2, 2A6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4 to reveal the potential of tubeimoside I to induce drug-drug interaction.The evaluation of cytochromes P450 enzyme (CYP) activity was performed in pooled human liver microsomes with probing substrates of CYP1A2, 2A6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4. Typical inhibitors were employed as positive controls and the effect of 0, 2.5, 5, 10, 25, 50, and 100 µM tubeimoside I was investigated.The activity of CYP2D6, 2E1, and 3A4 was significantly inhibited by tubeimoside I with the IC50 values of 10.34, 11.58, and 9.74 µM, respectively. The inhibition of CYP2D6 and 2E1 was competitive with the Ki value of 5.66 and 5.29 µM, respectively. While the inhibition of CYP3A4 was non-competitive with the Ki value of 4.87 µM. Moreover, the inhibition of CYP3A4 was time-dependent with the KI and Kinact values of 0.635 µM-1 and 0.0373 min-1, respectively.Tubeimoside I served as a competitive inhibitor of CYP2D6 and 2E1 exerting weak inhibition and a non-competitive inhibitor of CYP3A4 exerting moderate inhibition.


Subject(s)
Cytochrome P-450 CYP1A2 , Microsomes, Liver , Saponins , Triterpenes , Humans , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP2D6 , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/pharmacology
6.
Adv Healthc Mater ; 13(12): e2303699, 2024 May.
Article in English | MEDLINE | ID: mdl-38277695

ABSTRACT

Artificial cells are engineered units with cell-like functions for different purposes including acting as supportive elements for mammalian cells. Artificial cells with minimal liver-like function are made of alginate and equipped with metalloporphyrins that mimic the enzyme activity of a member of the cytochrome P450 family namely CYP1A2. The artificial cells are employed to enhance the dealkylation activity within 3D bioprinted structures composed of HepG2 cells and these artificial cells. This enhancement is monitored through the conversion of resorufin ethyl ether to resorufin. HepG2 cell aggregates are 3D bioprinted using an alginate/gelatin methacryloyl ink, resulting in the successful proliferation of the HepG2 cells. The composite ink made of an alginate/gelatin liquid phase with an increasing amount of artificial cells is characterized. The CYP1A2-like activity of artificial cells is preserved over at least 35 days, where 6 nM resorufin is produced in 8 h. Composite inks made of artificial cells and HepG2 cell aggregates in a liquid phase are used for 3D bioprinting. The HepG2 cells proliferate over 35 days, and the structure has boosted CYP1A2 activity. The integration of artificial cells and their living counterparts into larger 3D semi-synthetic tissues is a step towards exploring bottom-up synthetic biology in tissue engineering.


Subject(s)
Bioprinting , Cytochrome P-450 CYP1A2 , Printing, Three-Dimensional , Humans , Hep G2 Cells , Bioprinting/methods , Cytochrome P-450 CYP1A2/metabolism , Alginates/chemistry , Gelatin/chemistry , Tissue Engineering/methods , Cell Proliferation/drug effects , Metalloporphyrins/chemistry , Metalloporphyrins/pharmacology
7.
J Int Soc Sports Nutr ; 21(1): 2301384, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38226646

ABSTRACT

BACKGROUND: Caffeine (CAF) ingestion improves performance in a broad range of exercise tasks. Nevertheless, the CAF-induced, dose-dependent effect on discipline-specific performance and cognitive functions in CrossFit/High-Intensity Functional Training (HIFT) has not been sufficiently investigated. The aim of this study was to evaluate the effect of acute supplementation of three different doses of CAF and placebo (PLA) on specific performance, reaction time (RTime), postural stability (PStab), heart rate (HR) and perceived exertion (RPE). METHODS: In a randomized double-blind placebo-controlled crossover design, acute pre-exercise supplementation with CAF (3, 6, or 9 mg/kg body mass (BM)) and PLA in 26 moderately trained CrossFit practitioners was examined. The study protocol involved five separate testing sessions using the Fight Gone Bad test (FGB) as the exercise performance evaluation and biochemical analyses, HR and RPE monitoring, as well as the assessment of RTime and PStab, with regard to CYP1A2 (rs762551) and ADORA2A (rs5751876) single nucleotide polymorphism (SNP). RESULTS: Supplementation of 6 mgCAF/kgBM induced clinically noticeable improvements in FGBTotal results, RTime and pre-exercise motor time. Nevertheless, there were no significant differences between any CAF doses and PLA in FGBTotal, HRmax, HRmean, RPE, pre/post-exercise RTime, PStab variables or pyruvate concentrations. Lactate concentration was higher (p < 0.05) before and after exercise in all CAF doses than in PLA. There was no effect of CYP1A2 or ADORA2A SNPs on performance. CONCLUSIONS: The dose-dependent effect of CAF supplementation appears to be limited to statistically nonsignificant but clinically considered changes on specific performance, RTime, PStab, RPE or HR. However, regarding practical CAF-induced performance implications in CrossFit/HIFT, 6 mgCAF/kgBM may be supposed as the most rational supplementation strategy.


Subject(s)
Athletic Performance , Caffeine , Humans , Caffeine/pharmacology , Cross-Over Studies , Cytochrome P-450 CYP1A2 , Reaction Time , Athletic Performance/physiology , Lactic Acid , Double-Blind Method , Dietary Supplements , Polyesters
8.
Molecules ; 29(2)2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38276571

ABSTRACT

Human cytochrome P450 enzymes (CYPs) are critical for the metabolism of small-molecule pharmaceuticals (drugs). As such, the prediction of drug metabolism by and drug inhibition of CYP activity is an important component of the drug discovery and design process. Relative to the availability of a wide range of experimental atomic-resolution CYP structures, the development of structure-based CYP activity models has been limited. To better characterize the role of CYP conformational fluctuations in CYP activity, we perform multiple microsecond-scale all-atom explicit-solvent molecular dynamics (MD) simulations on three CYP isoforms, 1A2, 2D6, and 3A4, which together account for the majority of CYP-mediated drug metabolism. The MD simulations employ a variety of positional restraints, ranging from keeping all CYP atoms close to their experimentally determined coordinates to allowing full flexibility. We find that, with full flexibility, large fluctuations in the CYP binding sites correlate with efficient water exchange from these buried binding sites. This is especially true for 1A2, which, when restrained to its crystallographic conformation, is unable to exchange water between the binding site and bulk solvent. These findings imply that, in addition to crystal structures, a representative ensemble of conformational states ought to be included when developing structure-based CYP activity models.


Subject(s)
Cytochrome P-450 Enzyme System , Water , Humans , Water/metabolism , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 CYP1A2/metabolism , Binding Sites , Solvents , Microsomes, Liver/metabolism
9.
Eur J Hum Genet ; 32(3): 278-285, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37002327

ABSTRACT

The Dutch Pharmacogenetics Working Group (DPWG) aims to facilitate pharmacogenetics implementation in clinical practice by developing evidence-based guidelines to optimize pharmacotherapy. A guideline describing the gene-drug interaction between the genes CYP2D6, CYP3A4 and CYP1A2 and antipsychotics is presented here. The DPWG identified gene-drug interactions that require therapy adjustments when respective genotype is known for CYP2D6 with aripiprazole, brexpiprazole, haloperidol, pimozide, risperidone and zuclopenthixol, and for CYP3A4 with quetiapine. Evidence-based dose recommendations were obtained based on a systematic review of published literature. Reduction of the normal dose is recommended for aripiprazole, brexpiprazole, haloperidol, pimozide, risperidone and zuclopenthixol for CYP2D6-predicted PMs, and for pimozide and zuclopenthixol also for CYP2D6 IMs. For CYP2D6 UMs, a dose increase or an alternative drug is recommended for haloperidol and an alternative drug or titration of the dose for risperidone. In addition, in case of no or limited clinical effect, a dose increase is recommended for zuclopenthixol for CYP2D6 UMs. Even though evidence is limited, the DPWG recommends choosing an alternative drug to treat symptoms of depression or a dose reduction for other indications for quetiapine and CYP3A4 PMs. No therapy adjustments are recommended for the other CYP2D6 and CYP3A4 predicted phenotypes. In addition, no action is required for the gene-drug combinations CYP2D6 and clozapine, flupentixol, olanzapine or quetiapine and also not for CYP1A2 and clozapine or olanzapine. For identified gene-drug interactions requiring therapy adjustments, genotyping of CYP2D6 or CYP3A4 prior to treatment should not be considered for all patients, but on an individual patient basis only.


Subject(s)
Antipsychotic Agents , Clozapine , Quinolones , Thiophenes , Humans , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/pharmacology , Aripiprazole , Clopenthixol , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP3A/genetics , Drug Interactions , Haloperidol , Olanzapine , Pharmacogenetics , Pimozide , Quetiapine Fumarate/pharmacokinetics , Quetiapine Fumarate/pharmacology , Risperidone/pharmacokinetics , Risperidone/pharmacology
10.
Chem Biol Interact ; 387: 110811, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37993078

ABSTRACT

Ciprofol is a novel intravenous anesthetic agent. Its major glucuronide metabolite, M4, is found in plasma and urine. However, the specific isoforms of UDP-glucuronosyltransferases (UGTs) that metabolize ciprofol to M4 remain unknown. This study systematically characterized UGTs that contribute to the formation of M4 using human liver microsomes (HLM), human intestinal microsomes (HIM), and human recombinant UGTs. The inhibitory potential of ciprofol and M4 against major human UGTs and cytochrome P450 enzymes (P450s) was also explored. In vitro-in vivo extrapolation (IVIVE) and physiologically-based pharmacokinetic (PBPK) simulations were performed to predict potential in vivo drug-drug interactions (DDIs) caused by ciprofol. Glucuronidation of ciprofol followed Michaelis-Menten kinetics in both HLM and HIM with apparent Km values of 345 and 412 µM, Vmax values of 2214 and 444 nmol min-1·mg protein-1, respectively. The in vitro intrinsic clearances (CLint = Vmax/Km) for ciprofol glucuronidation by HLM and HIM were 6.4 and 1.1 µL min-1·mg protein-1, respectively. Human recombinant UGT studies revealed that UGT1A9 is the predominant isoform mediating M4 formation, followed by UGT1A7, with UGT1A8 playing a minor role. Ciprofol competitively inhibited CYP1A2 (Ki = 12 µM) and CYP2B6 (Ki = 4.7 µM), and noncompetitively inhibited CYP2C19 (Ki = 29 µM). No time-dependent inhibition by ciprofol was noted for CYP1A2, CYP2B6, or CYP2C19. In contrast, M4 showed limited or no inhibitory effects against selected P450s. Neither ciprofol nor M4 inhibited UGTs significantly. Initial IVIVE suggested potential ciprofol-mediated inhibition of CYP1A2, CYP2B6, and CYP2C19 inhibition in vivo. However, PBPK simulations showed no significant effect on phenacetin, bupropion, and S-mephenytoin exposure or peak plasma concentration. Our findings are pertinent for future DDI studies of ciprofol as either a perpetrator or victim drug.


Subject(s)
Cytochrome P-450 CYP1A2 , Microsomes, Liver , Humans , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C19/metabolism , Microsomes, Liver/metabolism , Glucuronosyltransferase/metabolism , Drug Interactions , Kinetics
11.
Toxicol Lett ; 391: 55-61, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38092155

ABSTRACT

This study investigates gene expression changes in laying hens exposed to trichothecene mycotoxins, known to induce oxidative stress and affect xenobiotic transformation and antioxidants. A 3-day feeding trial tested low and high doses of T-2/HT-2 toxin, DON/3-AcDON/15-AcDON, and FB1 in hen feed. Results showed increased expression of AHR, AHRR, HSP90, and CYP1A2 genes on days 2 and 3, suggesting a response to mycotoxin exposure. High doses down-regulated CYP1A2, AHR, and AHRR on day 1. KEAP1 expression decreased on day 1 but increased dose-dependently on days 2 and 3. NRF2 was up-regulated by low and down-regulated by high doses on day 1, then increased on days 2 and 3. Antioxidant-related genes (GPX3, GPX4, GSS, GSR) showed dose-dependent responses. Low doses up-regulated GPX3 and GPX4 throughout, while high doses up-regulated GPX3 on days 2 and 3 and GPX4 on day 3. GSS was up-regulated on day 3. Results indicate that toxic metabolites formed by phase I biotransformation rapidly induce ROS formation at low doses through the AHR/Hsp90/CYP1A2 pathway at the gene expression level, but at high levels, ROS-induced oxidative stress manifests later. Study showed simultaneous activation of redox-sensitive pathways: aryl hydrocarbon receptor (Ahr) and nuclear factor erythroid-derived 2-like 2 (Nrf2) by multi-mycotoxin exposure.


Subject(s)
Fusarium , Mycotoxins , T-2 Toxin , Female , Animals , Mycotoxins/toxicity , Fusarium/metabolism , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Chickens , Cytochrome P-450 CYP1A2/metabolism , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Antioxidants/metabolism , Liver/metabolism , T-2 Toxin/toxicity , T-2 Toxin/metabolism
12.
Free Radic Biol Med ; 211: 35-46, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38081439

ABSTRACT

Pregnant women exposed to polycyclic aromatic hydrocarbons (PAHs) are at increased risk for premature delivery. Premature infants often require supplemental oxygen, a known risk factor for bronchopulmonary dysplasia (BPD). Cytochrome P450 (CYP) enzymes have been implicated in hyperoxic lung injury. We hypothesize that prenatal PAH exposure exacerbates oxygen-mediated lung injury in neonatal mice, and that this effect is differentially altered in mice lacking the gene for (Cyp)1a1, 1a2, or 1b1. Timed pregnant wild type (WT) (C57BL/6J) mice were orally administered a PAH mixture of benzo[a]pyrene (BP) and benzo[b]fluoranthene (BbF) or the vehicle corn oil (CO) once daily on gestational days 16-19, and the dose response on postnatal lung injury was examined. In addition, timed pregnant mice with one of four genotypes, WT, Cyp1a1-null, Cyp1a2-null, and Cyp1b1-null, were treated orally with CO or PAH on gestational days 16-19 and exposed to hyperoxia or room air for 14 days. Lung injury was assessed on PND15 by radial alveolar count (RAC) and mean linear intercept (MLI) Gene expression of DNA repair genes in lung and liver were measured. Results showed that neonatal hyperoxic lung injury is augmented by prenatal PAH exposure in a dose-dependent manner. This effect was differentially altered in the Cyp-null mice, with Cyp1a2-null showing the greatest extent of lung injury. We concluded that newborn mice exposed to PAH in utero had more significant lung injury in response to hyperoxia than non-PAH exposed pups, and that CYP1A1 and CYP1A2 are protective against lung injury while CYP1B1 augments lung injury.


Subject(s)
Hyperoxia , Lung Injury , Polycyclic Aromatic Hydrocarbons , Prenatal Exposure Delayed Effects , Humans , Infant, Newborn , Female , Animals , Mice , Pregnancy , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Lung Injury/chemically induced , Hyperoxia/complications , Hyperoxia/genetics , Polycyclic Aromatic Hydrocarbons/toxicity , Polycyclic Aromatic Hydrocarbons/metabolism , Mice, Inbred C57BL , Lung/metabolism , Cytochrome P-450 Enzyme System , Oxygen , Mice, Knockout
13.
Br J Clin Pharmacol ; 90(1): 354-359, 2024 01.
Article in English | MEDLINE | ID: mdl-37596710

ABSTRACT

Clozapine (CLZ) is extensively used for treatment-resistant schizophrenia (TRS) with caution to avoid serious adverse events such as agranulocytosis and drug-drug interactions (DDIs). In the current report, we present a case of a 35-year-old male non-smoking TRS patient whose steady-state plasma trough concentrations (Ctrough ) of CLZ and its active metabolite, N-desmethylclozapine (NDMC), were significantly increased after initiating oral administration of lemborexant (LEM), a dual orexin receptor antagonist, for the treatment of insomnia. The patient experienced oversedation with sleepiness and fatigue while maintaining high levels of Ctrough of CLZ. The increased concentrations of CLZ returned to normal ranges after the discontinuation of LEM dosing, implying a pharmacokinetic DDI between CLZ and LEM. To gain insight into possible mechanisms, we performed in vitro assays of CYP1A2- and CYP3A4-mediated CLZ metabolism by measuring the formations of NDMC and clozapine N-oxide (CNO). In accordance with previous studies, the incubation of CLZ with each enzyme resulted in the production of both metabolites. LEM had only a weak inhibitory effect on CYP1A2- and CYP3A4-mediated CLZ metabolism. However, the preincubation of LEM with CYP3A4 in the presence of NADPH showed a significant enhancement of inhibitory effects on CLZ metabolism with IC50 values for the formations of CNO and NDMC of 2.8 µM and 4.1 µM, respectively, suggesting that LEM exerts as a potent time-dependent inhibitor for CYP3A4. Taken together, the results of the current study indicate that co-medication of CLZ with LEM may lead to increase in exposure to CLZ and risks of CLZ-related adverse events.


Subject(s)
Antipsychotic Agents , Clozapine , Male , Humans , Adult , Clozapine/adverse effects , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP3A/metabolism , Antipsychotic Agents/adverse effects , Drug Interactions
14.
Med Sci Sports Exerc ; 56(2): 328-339, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37844569

ABSTRACT

PURPOSE: This study aimed to summarize and meta-analyze existing evidence regarding the influence of CYP1A2 genotypes on the acute effects of caffeine for exercise performance and to investigate the interaction between genotype, dosage, and timing of caffeine supplementation. METHODS: Six databases were searched for studies determining the effect of caffeine (except mouth rinsing) on exercise performance between CYP1A2 genotypes. Three-level meta-analyses were performed using standardized mean differences (SMD; Hedge's g ) to determine the effect of caffeine on exercise outcomes within and between CYP1A2 genotypes (AA, AC, and CC). Meta-regressions were performed for dose, timing, and presence of reported conflict of interests (RCOI). A meta-analysis was also performed with placebo values to assess for imbalances between genotypes. RESULTS: Thirteen studies, totaling 119 outcomes and 440 participants, were included (233 AA, 175 AC, ad 34 CC). Caffeine improved performance for AA (SMD = 0.30, 95% confidence interval [CI] = 0.21-0.39, P < 0.0001) and AC (SMD = 0.16, 95% CI = 0.06-0.25, P = 0.022) but worsened performance for CC (SMD = -0.22, 95% CI = -0.44 to -0.01, P < 0.0001). Dose affected only CC, with greater doses generating more positive SMD (CC-dose estimate: +0.19/1 mg·kg -1 body mass, 95% CI = 0.04-0.33, P = 0.01). Timing influenced only CC, with better performance with later onset of exercise after supplementation (CC-timing estimate: +0.01/min, 95% CI = 0.00-0.02, P = 0.02). RCOI only affected SMD of CC (CC-RCOI estimate: -0.57, 95% CI = -1.02 to -0.12, P = 0.01). After excluding studies with RCOI, no influence of genotype was seen (all P ≥ 0.19). Small, nonsignificant differences were seen in placebo between genotypes (SMD AA vs CC: -0.13; AA vs AC: -0.12; AC vs CC: -0.05; all P ≥ 0.26). CONCLUSIONS: Caffeine improved performance for AA and AC but worsened performance for CC. Dose and timing moderated the efficacy of caffeine for CC only. Caution is advised because baseline differences and studies with RCOI could have influenced these results.


Subject(s)
Caffeine , Performance-Enhancing Substances , Humans , Caffeine/pharmacology , Cytochrome P-450 CYP1A2/genetics , Genotype , Exercise
15.
Eur J Epidemiol ; 39(1): 81-86, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37906419

ABSTRACT

Higher coffee consumption has been associated with reduced dementia risk, yet with inconsistencies across studies. CYP1A2 polymorphisms, which affects caffeine metabolism, may modulate the association between coffee and the risk of dementia and Alzheimer's disease (AD). We included 5964 participants of the Three-City Study (mean age 74 years-old), free of dementia at baseline when they reported their daily coffee consumption, with available genome-wide genotyping and followed for dementia over a median of 9.0 (range 0.8-18.7) years. In Cox proportional-hazards models, the relationship between coffee consumption and dementia risk was modified by CYP1A2 polymorphism at rs762551 (p for interaction = 0.034). In multivariable-adjusted models, coffee intake was linearly associated with a decreased risk of dementia among carriers of the C allele only ("slower caffeine metabolizers"; HR for 1-cup increased [95% CI] 0.90 [0.83-0.97]), while in non-carriers ("faster caffeine metabolizers"), there was no significant association but a J-shaped trend toward a decrease in dementia risk up to 3 cups/day and increased risk beyond. Thus, compared to null intake, drinking ≥ 4 cups of coffee daily was associated with a reduced dementia risk in slower but not faster metabolizers (HR [95% CI] for ≥ 4 vs. 0 cup/day = 0.45 [0.25-0.80] and 1.32 [0.89-1.96], respectively). Results were similar when studying AD and another CYP1A2 candidate polymorphism (rs2472304), but no interaction was found with CYP1A2 rs2472297 or rs2470893. In this cohort, a linear association of coffee intake to lower dementia risk was apparent only among carriers of CYP1A2 polymorphisms predisposing to slower caffeine metabolism.


Subject(s)
Coffee , Cytochrome P-450 CYP1A2 , Dementia , Aged , Humans , Caffeine/pharmacology , Caffeine/therapeutic use , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Dementia/epidemiology , Dementia/genetics , Risk Factors
16.
CPT Pharmacometrics Syst Pharmacol ; 13(3): 396-409, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38044486

ABSTRACT

Glofitamab is a novel T cell bispecific antibody developed for treatment of relapsed-refractory diffuse large B cell lymphoma and other non-Hodgkin's lymphoma indications. By simultaneously binding human CD20-expressing tumor cells and CD3 on T cells, glofitamab induces tumor cell lysis, in addition to T-cell activation, proliferation, and cytokine release. Here, we describe physiologically-based pharmacokinetic (PBPK) modeling performed to assess the impact of glofitamab-associated transient increases in interleukin 6 (IL-6) on the pharmacokinetics of several cytochrome P450 (CYP) substrates. By refinement of a previously described IL-6 model and inclusion of in vitro CYP suppression data for CYP3A4, CYP1A2, and 2C9, a PBPK model was established in Simcyp to capture the induced IL-6 levels seen when glofitamab is administered at the intended dose and dosing regimen. Following model qualification, the PBPK model was used to predict the potential impact of CYP suppression on exposures of various CYP probe substrates. PBPK analysis predicted that, in the worst-case, the transient elevation of IL-6 would increase exposures of CYP3A4, CYP2C9, and CYP1A2 substrates by less than or equal to twofold. Increases for CYP3A4, CYP2C9, and CYP1A2 substrates were projected to be 1.75, 1.19, and 1.09-fold following the first administration and 2.08, 1.28, and 1.49-fold following repeated administrations. It is recommended that there are no restrictions on concomitant treatment with any other drugs. Consideration may be given for potential drug-drug interaction during the first cycle in patients who are receiving concomitant CYP substrates with a narrow therapeutic index via monitoring for toxicity or for drug concentrations.


Subject(s)
Antibodies, Bispecific , Cytochrome P-450 CYP1A2 , Lymphoma, Non-Hodgkin , Humans , Interleukin-6 , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP2C9/metabolism , Drug Interactions , T-Lymphocytes/metabolism , Cytochrome P-450 Enzyme System/metabolism , Lymphoma, Non-Hodgkin/drug therapy , Models, Biological
17.
J Chem Inf Model ; 64(7): 2528-2538, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-37864562

ABSTRACT

Cytochrome P450 (CYP) is a family of enzymes that are responsible for about 75% of all metabolic reactions. Among them, CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 participate in the metabolism of most drugs and mediate many adverse drug reactions. Therefore, it is necessary to estimate the chemical inhibition of Cytochrome P450 enzymes in drug discovery and the food industry. In the past few decades, many computational models have been reported, and some provided good performance. However, there are still several issues that should be resolved for these models, such as single isoform, models with unbalanced performance, lack of structural characteristics analysis, and poor availability. In the present study, the deep learning models based on python using the Keras framework and TensorFlow were developed for the chemical inhibition of each CYP isoform. These models were established based on a large data set containing 85715 compounds extracted from the PubChem bioassay database. On external validation, the models provided good AUC values with 0.97, 0.94, 0.94, 0.96, and 0.94 for CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4, respectively. The models can be freely accessed on the Web server named CYPi-DNNpredictor (cypi.sapredictor.cn), and the codes for the model were made open source in the Supporting Information. In addition, we also analyzed the structural characteristics of chemicals with CYP450 inhibition and detected the structural alerts (SAs), which should be responsible for the inhibition. The SAs were also made available online, named CYPi-SAdetector (cypisa.sapredictor.cn). The models can be used as a powerful tool for the prediction of CYP450 inhibitors, and the SAs should provide useful information for the mechanisms of Cytochrome P450 inhibition.


Subject(s)
Cytochrome P-450 CYP1A2 , Deep Learning , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6 , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP2C9 , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Protein Isoforms , Microsomes, Liver/metabolism
19.
Bipolar Disord ; 26(1): 95-97, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38097824

ABSTRACT

We report the case of a Chinese male with schizoaffective disorder, an active smoker and a nonresponder to clozapine (600 mg daily). Therapeutic clozapine monitoring was analyzed, revealing a low concentration-dose ratio. A pharmacogenetic test showed that the patient had the CYP1A2*1F/*1F genotype, indicating an ultra-rapid clozapine metabolizer. In combination with fluvoxamine, a CYP1A2 enzyme inhibitor, clozapine plasma concentrations approached the reference range and achieved clinical improvement. This case demonstrates how pharmacogenetics can help understand the value of therapeutic drug monitoring to enhance the treatment of refractory schizoaffective disorder.


Subject(s)
Antipsychotic Agents , Bipolar Disorder , Clozapine , Psychotic Disorders , Male , Humans , Clozapine/therapeutic use , Cytochrome P-450 CYP1A2/genetics , Antipsychotic Agents/therapeutic use , Bipolar Disorder/drug therapy , Psychotic Disorders/drug therapy , Psychotic Disorders/genetics , Genetic Testing
20.
Med Res Rev ; 44(1): 169-234, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37337403

ABSTRACT

Mammalian cytochrome P450 1A (CYP1A) are key phase I xenobiotic-metabolizing enzymes that play a distinctive role in metabolic activation or metabolic clearance of a variety of procarcinogens, drugs, and endogenous substances. Human CYP1A subfamily contains two members (hCYP1A1 and hCYP1A2), which are known to catalyze the oxidative activation of some environmental procarcinogens into carcinogenic species. Increasing evidence has demonstrated that CYP1A inhibitor therapies are promising strategies for cancer chemoprevention or overcoming CYP1A-associated drug toxicity and resistance. Herein, we reviewed recent advances in the discovery and characterization of hCYP1A inhibitors, from the discovery approaches to structural features and biomedical applications of hCYP1A inhibitors. The inhibition potentials, inhibition modes, and inhibition constants of all reported hCYP1A inhibitors are comprehensively summarized. Meanwhile, the structural features and structure-activity relationships of different classes of hCYP1A1 and hCYP1A2 inhibitors are analyzed and discussed in depth. Furthermore, the major challenges and future directions for this field are presented and highlighted. Collectively, the information and knowledge presented here will strongly facilitate the researchers to discover and develop more efficacious CYP1A inhibitors for specific purposes, such as chemo-preventive agents or as tool molecules in hCYP1A-related fundamental studies.


Subject(s)
Cytochrome P-450 CYP1A1 , Cytochrome P-450 CYP1A2 , Animals , Humans , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 Enzyme System/metabolism , Mammals/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...