Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.736
Filter
1.
Yakugaku Zasshi ; 144(7): 775-779, 2024.
Article in English | MEDLINE | ID: mdl-38945852

ABSTRACT

Venetoclax (VEN) is used in patients with acute myeloid leukemia (AML) and is primarily metabolized by CYP3A4, a major drug-metabolizing enzyme. Patients with AML simultaneously administered VEN and CYP3A4 inhibitors require a more appropriate management of drug-drug interactions (DDIs). Here, we report two cases of patients with AML (54-year-old man and 22-year-old woman) administrated VEN and CYP3A4 inhibitors, such as posaconazole, cyclosporine, or danazol. In the first case, we evaluated the appropriateness of timing for adjusting VEN dosage subsequent to the cessation of posaconazole. Consequently, modifying the VEN dosage in conjunction with the cessation of Posaconazole simultaneously may result in elevated plasma VEN levels. In the second case, plasma VEN concentrations were markedly elevated when co-administered with several CYP3A4 inhibitors. Additionally, in vitro assays were conducted for reverse translational studies to analyze CYP3A4 inhibition. CYP3A4 inhibition by combinatorial administration of cyclosporine A and danazol was demonstrated in vitro, which potentially explains the increasing plasma VEN concentrations observed in clinical settings. Although the acquisition of therapeutic effects is a major priority for patients, frequent therapeutic drug monitoring and dosage adjustments considering DDIs would be important factors in chemotherapy.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A , Drug Interactions , Drug Monitoring , Leukemia, Myeloid, Acute , Sulfonamides , Humans , Sulfonamides/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Male , Young Adult , Middle Aged , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/blood , Female , Cytochrome P-450 CYP3A/metabolism , Cyclosporine/administration & dosage , Triazoles/administration & dosage , Antineoplastic Agents/administration & dosage
2.
Ann Acad Med Singap ; 53(2): 69-79, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38920231

ABSTRACT

Introduction: Few real-world studies have investigated drug-drug interactions (DDIs) involving non-vitamin-K antagonist oral anticoagulants (NOACs) in patients with nonvalvular atrial fibrillation (NVAF). The interactions encompass drugs inducing or inhibiting cytochrome P450 3A4 and permeability glycoprotein. These agents potentially modulate the breakdown and elimination of NOACs. This study investigated the impact of DDIs on thromboembolism in this clinical scenario. Method: Patients who had NVAF and were treated with NOACs were selected as the study cohort from the National Health Insurance Research Database of Taiwan. Cases were defined as patients hospitalised for a thromboembolic event and who underwent a relevant imaging study within 7 days before hospitalisa-tion or during hospitalisation. Each case was matched with up to 4 controls by using the incidence density sampling method. The concurrent use of a cytochrome P450 3A4/permeability glycoprotein inducer or inhibitor or both with NOACs was identified. The effects of these interactions on the risk of thromboembolic events were examined with univariate and multivariate conditional logistic regressions. Results: The study cohort comprised 60,726 eligible patients. Among them, 1288 patients with a thromboembolic event and 5144 matched control patients were selected for analysis. The concurrent use of a cytochrome P450 3A4/permeability glycoprotein inducer resulted in a higher risk of thromboembolic events (adjusted odds ratio [AOR] 1.23, 95% confidence interval [CI] 1.004-1.51). Conclusion: For patients with NVAF receiving NOACs, the concurrent use of cytochrome P450 3A4/ permeability glycoprotein inducers increases the risk of thromboembolic events.


Subject(s)
Anticoagulants , Atrial Fibrillation , Drug Interactions , Thromboembolism , Humans , Atrial Fibrillation/drug therapy , Atrial Fibrillation/complications , Thromboembolism/prevention & control , Thromboembolism/epidemiology , Thromboembolism/etiology , Anticoagulants/administration & dosage , Anticoagulants/therapeutic use , Male , Female , Aged , Administration, Oral , Taiwan/epidemiology , Middle Aged , Case-Control Studies , Aged, 80 and over , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A/metabolism , Factor Xa Inhibitors/therapeutic use , Factor Xa Inhibitors/administration & dosage , Pyridones/administration & dosage , Pyridones/therapeutic use , Pyridones/adverse effects
3.
Toxicol Lett ; 397: 34-41, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38734219

ABSTRACT

Humantenmine, koumine, and gelsemine are three indole alkaloids found in the highly toxic plant Gelsemium. Humantenmine was the most toxic, followed by gelsemine and koumine. The aim of this study was to investigate and analyze the effects of these three substances on tissue distribution and toxicity in mice pretreated with the Cytochrome P450 3A4 (CYP3A4) inducer ketoconazole and the inhibitor rifampicin. The in vivo test results showed that the three alkaloids were absorbed rapidly and had the ability to penetrate the blood-brain barrier. At 5 min after intraperitoneal injection, the three alkaloids were widely distributed in various tissues and organs, the spleen and pancreas were the most distributed, and the content of all tissues decreased significantly at 20 min. Induction or inhibition of CYP3A4 in vivo can regulate the distribution and elimination effects of the three alkaloids in various tissues and organs. Additionally, induction of CYP3A4 can reduce the toxicity of humantenmine, and vice versa. Changes in CYP3A4 levels may account for the difference in toxicity of humantenmine. These findings provide a reliable and detailed dataset for drug interactions, tissue distribution, and toxicity studies of Gelsemium alkaloids.


Subject(s)
Cytochrome P-450 CYP3A , Gelsemium , Indole Alkaloids , Animals , Gelsemium/chemistry , Cytochrome P-450 CYP3A/metabolism , Indole Alkaloids/toxicity , Tissue Distribution , Male , Mice , Ketoconazole/toxicity , Ketoconazole/pharmacology , Cytochrome P-450 CYP3A Inducers/pharmacology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Alkaloids
4.
J Int Med Res ; 52(5): 3000605241247705, 2024 May.
Article in English | MEDLINE | ID: mdl-38698526

ABSTRACT

Nirmatrelvir/ritonavir is a novel drug combination that is authorized by the Food and Drug Administration for the treatment of coronavirus disease 2019 (COVID-19). Ritonavir is a cytochrome P450 3A inhibitor and a P-glycoprotein inhibitor that increases the plasma concentration of tacrolimus and other medications. We describe the cases of two patients treated with nirmatrelvir/ritonavir: a patient who had undergone kidney transplantation and another with a history of hematopoietic stem cell transplantation. Toxic concentrations of tacrolimus were induced in both. This case series highlights the risk associated with the concomitant administration of tacrolimus and nirmatrelvir/ritonavir.


Subject(s)
COVID-19 Drug Treatment , Drug Interactions , Kidney Transplantation , Ritonavir , Tacrolimus , Humans , Ritonavir/therapeutic use , Tacrolimus/therapeutic use , Tacrolimus/adverse effects , Male , Middle Aged , SARS-CoV-2/isolation & purification , Female , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Cytochrome P-450 CYP3A Inhibitors/therapeutic use , Drug Combinations , COVID-19/virology , Aged , Antiviral Agents/therapeutic use
5.
Pharmacotherapy ; 44(6): 480-484, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38819020

ABSTRACT

BACKGROUND: Intranasal esketamine is an approved drug for treatment­resistant depression (TRD); however, it is costly and may result in specific adverse effects. In this single case study, we explored if oral esketamine can be a suitable alternative. METHODS: In collaboration with a 39­year­old female with TRD, we compared plasma concentration curves of intranasal (84 mg) and oral (1, 2 and 4 mg/kg) esketamine. Because oral esketamine has a relatively low bioavailability, it results in a different ratio between esketamine and its primary metabolite noresketamine. To increase the bioavailability of oral esketamine, we co­administered a single dose of the cytochrome P­450 (CYP) 3A4 inhibitor cobicistat (150 mg). RESULTS: For all doses administered, oral esketamine resulted in lower esketamine but higher noresketamine peak plasma concentrations compared with intranasal treatment. Using oral esketamine it was not possible to generate a similar esketamine plasma concentration curve as with the intranasal treatment, except when combined with cobicistat (esketamine 2 mg/kg plus cobicistat 150 mg). CONCLUSIONS: Our findings demonstrate that cobicistat effectively increases the bioavailability of oral esketamine. Further research is required in a larger population, especially to investigate the clinical benefit of cobicistat as a booster drug for oral esketamine.


Subject(s)
Administration, Intranasal , Biological Availability , Cobicistat , Depressive Disorder, Treatment-Resistant , Ketamine , Ketamine/administration & dosage , Ketamine/pharmacokinetics , Female , Humans , Adult , Administration, Oral , Cobicistat/administration & dosage , Cobicistat/pharmacokinetics , Depressive Disorder, Treatment-Resistant/drug therapy , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions
6.
Eur J Med Chem ; 273: 116492, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38762918

ABSTRACT

Paclitaxel (PTX) is considered the blockbuster chemotherapy treatment for cancer. Paclitaxel's (PTX) oral administration has proven to be extremely difficult, mostly because of its susceptibility to intestinal P-glycoprotein (P-gp) and cytochrome P450 (CYP3A4). The concurrent local inhibition of intestinal P-gp and CYP3A4 is a promising approach to improve the oral bioavailability of paclitaxel while avoiding potential unfavorable side effects of their systemic inhibition. Herein, we report the rational design and evaluation of novel dual potent inhibitors of P-gp and CYP3A4 using an anthranilamide derivative tariquidar as a starting point for their structural optimizations. Compound 14f, bearing N-imidazolylbenzyl side chain, was found to have potent and selective P-gp (EC50 = 28 nM) and CYP3A4 (IC50 = 223 nM) inhibitory activities with low absorption potential (Papp (A-to-B) <0.06). In vivo, inhibitor 14f improved the oral absorption of paclitaxel by 6 times in mice and by 30 times in rats as compared to vehicle, while 14f itself remained poorly absorbed. Compound 14f, possessing dual P-gp and CYP3A4 inhibitory activities, offered additional enhancement in paclitaxel oral absorption compared to tariquidar in mice. Evaluating the CYP effect of 14f on oral absorption of paclitaxel requires considering the variations in CYP expression between animal species. This study provides further medicinal chemistry advice on strategies for resolving concerns with the oral administration of chemotherapeutic agents.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A , Drug Design , ortho-Aminobenzoates , Cytochrome P-450 CYP3A/metabolism , Humans , Animals , ortho-Aminobenzoates/pharmacology , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/chemical synthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Mice , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/chemical synthesis , Cytochrome P-450 CYP3A Inhibitors/chemistry , Structure-Activity Relationship , Molecular Structure , Models, Molecular , Rats , Dose-Response Relationship, Drug , Paclitaxel/pharmacology , Paclitaxel/chemistry , Male
7.
Drug Metab Pharmacokinet ; 56: 101005, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663182

ABSTRACT

We previously reported novel benzyl-ether derivatives with an imidazole ring and a hydroxyl group (A-01) or carboxyl group (B-01) and esters (2 esters of A-01, and 7 esters of B-01) as pharmacokinetics (PK) boosters. This study demonstrates how these ester compounds embody the concept of a safe pharmacokinetic booster, with potent and transient inhibition of CYP3A4-mediated drug metabolism. As a model CYP3A4 substrate and CYP3A4 enzyme, midazolam (MDZ) and rat liver microsomes were used. A-01 inhibited MDZ metabolism significantly, while B-01 induced only slight inhibition. Although rat liver microsomes hydrolyzed the ester compounds over time, several ester compounds strongly inhibited MDZ metabolism. Due to the significant activity of A-01, A-01 esters affected MDZ metabolism, irrespective of hydrolysis state. Time-dependent inhibition evaluation indicated that the B-01 ester inhibition is not mechanism-based, as hydrolysis eliminated MDZ metabolism inhibition. We report that the B-01 esters significantly inhibit CYP3A4-mediated drug metabolism, and upon hydrolysis this property is eliminated. In conclusion, B-01 ester compounds may be safe PK boosters with antedrug characteristics.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A , Microsomes, Liver , Midazolam , Animals , Cytochrome P-450 CYP3A/metabolism , Microsomes, Liver/metabolism , Midazolam/metabolism , Midazolam/pharmacokinetics , Rats , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Male , Rats, Sprague-Dawley , Esters/chemistry , Esters/metabolism
8.
Int J Biol Macromol ; 268(Pt 2): 131502, 2024 May.
Article in English | MEDLINE | ID: mdl-38626834

ABSTRACT

Piperlonguminine (PLG) is a major alkaloid found in Piper longum fruits. It has been shown to possess a variety of biological activities, including anti-tumor, anti-hyperlipidemic, anti-renal fibrosis and anti-inflammatory properties. Previous studies have reported that PLG inhibits various CYP450 enzymes. The main objective of this study was to identify reactive metabolites of PLG in vitro and assess its ability to inhibit CYP450. In rat and human liver microsomal incubation systems exposed to PLG, two oxidized metabolites (M1 and M2) were detected. Additionally, in microsomes where N-acetylcysteine was used as a trapping agent, N-acetylcysteine conjugates (M3, M4, M5 and M6) of four isomeric O-quinone-derived reactive metabolites were found. The formation of metabolites was dependent on NADPH. Inhibition and recombinant CYP450 enzyme incubation experiments showed that CYP3A4 was the primary enzyme responsible for the metabolic activation of PLG. This study characterized the O-dealkylated metabolite (M1) through chemical synthesis. The IC50 shift assay showed time-dependent inhibition of CYP3A4, 2C9, 2E1, 2C8 and 2D6 by PLG. This research contributes to the understanding of PLG-induced enzyme inhibition and bioactivation.


Subject(s)
Activation, Metabolic , Cytochrome P-450 CYP3A , Dioxolanes , Microsomes, Liver , Animals , Humans , Cytochrome P-450 CYP3A/metabolism , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects , Rats , Dioxolanes/pharmacology , Dioxolanes/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Male , Piperidones , Benzodioxoles
9.
Mar Drugs ; 22(4)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38667795

ABSTRACT

This open-label, two-part, phase Ib drug-drug interaction study investigated whether the pharmacokinetic (PK) and safety profiles of lurbinectedin (LRB), a marine-derived drug, are affected by co-administration of itraconazole (ITZ), a strong CYP3A4 inhibitor, in adult patients with advanced solid tumors. In Part A, three patients were sequentially assigned to Sequence 1 (LRB 0.8 mg/m2, 1-h intravenous [IV] + ITZ 200 mg/day oral in Cycle 1 [C1] and LRB alone 3.2 mg/m2, 1 h, IV in Cycle 2 [C2]). In Part B, 11 patients were randomized (1:1) to receive either Sequence 1 (LRB at 0.9 mg/m2 + ITZ in C1 and LRB alone in C2) or Sequence 2 (LRB alone in C1 and LRB + ITZ in C2). Eleven patients were evaluable for PK analysis: three in Part A and eight in Part B (four per sequence). The systemic total exposure of LRB increased with ITZ co-administration: 15% for Cmax, area under the curve (AUC) 2.4-fold for AUC0-t and 2.7-fold for AUC0-∞. Co-administration with ITZ produced statistically significant modifications in the unbound plasma LRB PK parameters. The LRB safety profile was consistent with the toxicities described in previous studies. Co-administration with multiple doses of ITZ significantly altered LRB systemic exposure. Hence, to avoid LRB overexposure when co-administered with strong CYP3A4 inhibitors, an LRB dose reduction proportional to CL reduction should be applied.


Subject(s)
Carbolines , Cytochrome P-450 CYP3A Inhibitors , Drug Interactions , Heterocyclic Compounds, 4 or More Rings , Itraconazole , Neoplasms , Humans , Itraconazole/pharmacokinetics , Itraconazole/administration & dosage , Itraconazole/adverse effects , Male , Middle Aged , Female , Aged , Neoplasms/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Heterocyclic Compounds, 4 or More Rings/adverse effects , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Carbolines/pharmacokinetics , Carbolines/administration & dosage , Carbolines/adverse effects , Adult , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/adverse effects , Area Under Curve , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/administration & dosage
10.
Curr Pharm Des ; 30(15): 1167-1177, 2024.
Article in English | MEDLINE | ID: mdl-38523519

ABSTRACT

BACKGROUND: Metabolism of oral anticoagulants (OAC) is affected by P-glycoprotein (P-gp)/ CYP3A4 enzyme. However, the P-gp/CYP3A4 inhibitors are unavoidably used with OACs. METHODS: Medline, Cochrane, and Embase were systematically searched for randomized controlled trials and cohort studies from inception till 23rd November, 2022 to assess the safety and effectiveness of OACs when concomitantly used with P-gp/CYP3A4 inhibitors. The primary outcomes were major bleeding and gastrointestinal (GI) bleeding. Secondary outcomes were stroke/systemic embolism (SE), all-cause mortality, any bleeding as well as intracranial hemorrhage (ICH). We estimated summary odds ratios (OR) with 95% credible intervals (CI) using pairwise and network meta-analysis with random effects. RESULTS: A total of 11 studies involving 37,973 patients were included. When concomitantly used with P-pg/ CYP3A4 inhibitors, network meta-analysis indicated that dabigatran, apixaban, and edoxaban were associated with significantly lower risk of major bleeding compared to rivaroxaban, with ORs of 0.56, 0.51 and 0.48, respectively. Rivaroxaban and dabigatran were associated with a significantly increased risk of GI bleeding than warfarin, apixaban and edoxaban. Dabigatran and apixaban were linked with significantly lower risk of any bleeding compared with warfarin (ORs were 0.75 and 0.68, respectively) or rivaroxaban (ORs were 0.67 and 0.60, respectively). Apixaban (OR 0.32) and edoxaban (OR 0.35) were associated with a lower risk of ICH compared with warfarin. There was no difference between any OACs in terms of stroke/SE or all-cause mortality. CONCLUSION: When concomitantly used with P-gp/CYP3A4 inhibitors, apixaban and edoxaban were associated with a lower risk of bleeding, though no significant difference in effectiveness was observed among all OACs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Anticoagulants , Cytochrome P-450 CYP3A , Humans , Anticoagulants/adverse effects , Anticoagulants/administration & dosage , Cytochrome P-450 CYP3A/metabolism , Administration, Oral , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Network Meta-Analysis , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Hemorrhage/chemically induced
11.
CPT Pharmacometrics Syst Pharmacol ; 13(4): 660-672, 2024 04.
Article in English | MEDLINE | ID: mdl-38481038

ABSTRACT

Pralsetinib, a potent and selective inhibitor of oncogenic RET fusion and RET mutant proteins, is a substrate of the drug metabolizing enzyme CYP3A4 and a substrate of the efflux transporter P-gp based on in vitro data. Therefore, its pharmacokinetics (PKs) may be affected by co-administration of potent CYP3A4 inhibitors and inducers, P-gp inhibitors, and combined CYP3A4 and P-gp inhibitors. With the frequent overlap between CYP3A4 and P-gp substrates/inhibitors, pralsetinib is a challenging and representative example of the need to more quantitatively characterize transporter-enzyme interplay. A physiologically-based PK (PBPK) model for pralsetinib was developed to understand the victim drug-drug interaction (DDI) risk for pralsetinib. The key parameters driving the magnitude of pralsetinib DDIs, the P-gp intrinsic clearance and the fraction metabolized by CYP3A4, were determined from PBPK simulations that best captured observed DDIs from three clinical studies. Sensitivity analyses and scenario simulations were also conducted to ensure these key parameters were determined with sound mechanistic rationale based on current knowledge, including the worst-case scenarios. The verified pralsetinib PBPK model was then applied to predict the effect of other inhibitors and inducers on the PKs of pralsetinib. This work highlights the challenges in understanding DDIs when enzyme-transporter interplay occurs, and demonstrates an important strategy for differentiating enzyme/transporter contributions to enable PBPK predictions for untested scenarios and to inform labeling.


Subject(s)
Cytochrome P-450 CYP3A , Pyrazoles , Pyrimidines , Humans , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Pyridines , Membrane Transport Proteins , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Models, Biological
12.
J Thromb Thrombolysis ; 57(4): 598-602, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38554223

ABSTRACT

Moderate-strong CYP3A4 or Pgp inhibitors and inducers alter direct oral anticoagulant (DOAC) pharmacokinetics. Whether the presence of a DOAC drug-drug interaction (DDI) prompts in- hospital changes in management remains unknown. We identified all hospitalized patients at our institution who were admitted with a clinically relevant DOAC DDI from 01/2021 to 06/2021. Clinically relevant DOAC DDIs were defined as those listed in the prescribing information or FDA CYP3A4/Pgp inhibitors clinical indexes. We assessed the prevalence of DOAC DDIs and categorized their management as: drug stopped, drug held, or drug continued. For drugs that were continued we assessed whether the dose of the DOAC or interacting drug was increased, decreased or unchanged during the admission. We ascertained the number of DOAC DDIs that prompted an automated prescribing alert in our electronic health record (EHR). Finally, we conducted a logistic regression model to compare users of DOACs with DDI who had their regimen adjusted versus those without adjustments, focusing on outcomes of rehospitalization and death, adjusting for age and gender. Among 3,725 hospitalizations with a DOAC admission order, 197 (5%) had a clinically relevant DOAC DDI. The DOAC and the interacting drug were continued at discharge for 124 (63%) hospitalizations. The most frequent adjustments were stopping the interacting drug (73%) and stopping the DOAC (15%). Only 7 (4%) of DOAC DDIs prompted an EHR alert. The adjusted odds ratios for rehospitalizations and death, respectively, among patients whose regimens were adjusted compared to those whose were not, were 1.29 (95% CI, 0.67 to 2.48; P = 0.44) and 1.88 (95% CI, 0.91 to 3.89; P = 0.09). Clinically relevant DDIs with DOACs occur infrequently among hospitalized patients and usually are managed without stopping the DOAC. The clinical impact of such DDIs and subsequent adjustments on thrombotic and hemorrhagic outcomes requires further investigation.


Subject(s)
Cytochrome P-450 CYP3A , Hemorrhage , Humans , Drug Interactions , Hemorrhage/drug therapy , Cytochrome P-450 CYP3A Inhibitors , Anticoagulants/therapeutic use , Administration, Oral
13.
Drug Metab Pharmacokinet ; 55: 101000, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38458122

ABSTRACT

In this study, a physiologically based pharmacokinetic (PBPK) model of the cytochrome P450 3A (CYP3A) substrate azelnidipine was developed using in vitro and clinical data to predict the effects of azole antifungals on azelnidipine pharmacokinetics. Modeling and simulations were conducted using the Simcyp™ PBPK simulator. The azelnidipine model consisted of a full PBPK model and a first-order absorption model. CYP3A was assumed as the only azelnidipine elimination route, and CYP3A clearance was optimized using the pharmacokinetic profile of single-dose 5-mg azelnidipine in healthy participants. The model reproduced the results of a clinical drug-drug interaction study and met validation criteria. PBPK model simulations using azole antifungals (itraconazole, voriconazole, posaconazole, fluconazole, fosfluconazole) and azelnidipine or midazolam (CYP3A index substrate) were performed. Increases in the simulated area under the plasma concentration-time curve from time zero extrapolated to infinity with inhibitors were comparable between azelnidipine (range, 2.11-6.47) and midazolam (range, 2.26-9.22), demonstrating that azelnidipine is a sensitive CYP3A substrate. Increased azelnidipine plasma concentrations are expected when co-administered with azole antifungals, potentially affecting azelnidipine safety. These findings support the avoidance of azole antifungals in patients taking azelnidipine and demonstrate the utility of PBPK modeling to inform appropriate drug use.


Subject(s)
Antifungal Agents , Azetidinecarboxylic Acid/analogs & derivatives , Dihydropyridines , Midazolam , Humans , Midazolam/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A , Drug Interactions , Itraconazole , Models, Biological
14.
J Med Chem ; 67(4): 2802-2811, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38330258

ABSTRACT

Chelerythrine chloride (CHE) is a quaternary benzo[c]phenanthridine alkaloid with an iminium group that was found to cause time- and concentration-dependent inhibition of CYP3A4. The loss of CYP3A4 activity was independent of NADPH. CYP3A4 competitive inhibitor ketoconazole and nucleophile N-acetylcysteine (NAC) slowed the inactivation. No recovery of CYP3A4 activity was observed after dialysis. Dihydrochelerythrine hardly inhibited CYP3A4, suggesting that the iminium group was primarily responsible for the inactivation. UV spectral analysis revealed that the maximal absorbance of CHE produced a significant red-shift after being mixed with NAC, suggesting that 1,2-addition possibly took place between the sulfhydryl group of NAC and iminium group of CHE. Molecular dynamics simulation and site-direct mutagenesis studies demonstrated that modification of Cys239 by the iminium group of CHE attributed to the inactivation. In conclusion, CHE is an affinity-labeling inactivator of CYP3A4. The observed enzyme inactivation resulted from the modification of Cys239 of CYP3A4 by the iminium group of CHE.


Subject(s)
Alkaloids , Antineoplastic Agents , Benzophenanthridines , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors/pharmacology
15.
Clin Pharmacol Drug Dev ; 13(7): 810-818, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38421129

ABSTRACT

Sotorasib is a small molecule that irreversibly inhibits the Kirsten rat sarcoma viral oncogene homolog (KRAS) protein with a G12C amino acid substitution mutant protein. The impact of cytochrome P450 (CYP) 3A4 inhibition and induction on sotorasib pharmacokinetics (PKs) was evaluated in 2 separate studies in healthy volunteers (N = 14/study). The impact of CYP3A4 inhibition was interrogated utilizing repeat doses of 200 mg of itraconazole, a strong CYP3A4 inhibitor, on 360 mg of sotorasib PKs. The impact of CYP3A4 induction was interrogated utilizing multiple doses of 600 mg of rifampin, a strong CYP3A4 inducer. Additionally, the impact of organic anion transporting polypeptide (OATP) 1B1/3 inhibition on 960 mg of sotorasib PKs was interrogated after a single dose of 600 mg of rifampin. CYP3A4 inhibition did not significantly impact sotorasib Cmax but did lead to a 26% increase in sotorasib AUCinf. CYP3A4 induction decreased sotorasib Cmax by 35% and AUCinf by 51%. OATP1B1/3 inhibition decreased sotorasib Cmax and AUCinf by 16% and 23%, respectively. These results support that sotorasib can be given together with strong CYP3A4 and OATP1B1/3 inhibitors but the co-administration of sotorasib and strong CYP3A4 inducers should be avoided.


Subject(s)
Cytochrome P-450 CYP3A Inducers , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A , Drug Interactions , Liver-Specific Organic Anion Transporter 1 , Rifampin , Solute Carrier Organic Anion Transporter Family Member 1B3 , Humans , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Adult , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A/metabolism , Rifampin/pharmacology , Rifampin/administration & dosage , Cytochrome P-450 CYP3A Inducers/pharmacology , Female , Young Adult , Administration, Oral , Middle Aged , Healthy Volunteers , Area Under Curve , Itraconazole/pharmacology , Itraconazole/administration & dosage , Itraconazole/pharmacokinetics , Spiro Compounds/pharmacokinetics , Spiro Compounds/administration & dosage , Spiro Compounds/pharmacology
16.
Clin Ther ; 46(3): 194-200, 2024 03.
Article in English | MEDLINE | ID: mdl-38307724

ABSTRACT

PURPOSE: Trofinetide is the first drug to be approved by the US Food and Drug Administration for use in the treatment of patients with Rett syndrome, a multisystem disorder requiring multimodal therapies. Cytochrome P450 (CYP) 3A4 metabolizes >50% of therapeutic drugs and is the CYP isozyme most commonly expressed in the liver and intestines. In vitro studies suggest the concentration of trofinetide producing 50% inhibition (IC50) of CYP3A4 is >15 mmol/L; that concentration was much greater than the target clinical concentration associated with the maximal intended therapeutic dose (12 g). Thus, trofinetide has a low potential for drug-drug interactions in the liver. However, there is potential for drug-drug interactions in the intestines given the oral route of administration and expected relatively high concentration in the gastrointestinal tract after dose administration. METHODS: Using a validated physiologically based pharmacokinetic (PBPK) model, deterministic and stochastic simulations were used for assessing the PK properties related to exposure and bioavailability of midazolam (sensitive index substrate for CYP3A4) following an oral (15 mg) or intravenous (2 mg) dose, with and without single-dose and steady-state (12 g) coadministration of oral trofinetide. FINDINGS: Following coadministration of intravenous midazolam and oral trofinetide, the PK properties of midazolam were unchanged. The trofinetide concentration in the gut wall was >15 mmol/L during the first 1.5 hours after dosing. With the coadministration of oral midazolam and trofinetide, the model predicted increases in fraction of dose reaching the portal vein, bioavailability, Cmax, and AUCinf of 30%, 30%, 18%, and 30%, respectively. IMPLICATIONS: In this study that used a PBPK modeling approach, it was shown that CYP3A4 enzyme activity in the liver was not affected by trofinetide coadministration, but trofinetide was predicted to be a weak inhibitor of intestinal CYP3A4 metabolism after oral administration at therapeutic doses.


Subject(s)
Cytochrome P-450 CYP3A , Glutamates , Midazolam , Humans , Pharmaceutical Preparations , Cytochrome P-450 CYP3A/metabolism , Midazolam/pharmacokinetics , Drug Interactions , Models, Biological , Cytochrome P-450 CYP3A Inhibitors
17.
AAPS J ; 26(2): 26, 2024 02 16.
Article in English | MEDLINE | ID: mdl-38366061

ABSTRACT

CYP3A is one of the most important classes of enzymes and is involved in the metabolism of over 70% drugs. While several selective CYP3A4 inhibitors have been identified, the search for a selective CYP3A5 inhibitor has turned out to be rather challenging. Recently, several selective CYP3A5 inhibitors have been identified through high-throughput screening of ~ 11,000 compounds and hit expansion using human recombinant enzymes. We set forth to characterize the three most selective CYP3A5 inhibitors in a more physiologically relevant system of human liver microsomes to understand if these inhibitors can be used for reaction phenotyping studies in drug discovery settings. Gomisin A and T-5 were used as selective substrate reactions for CYP3A4 and CYP3A5 to determine IC50 values of the two enzymes. The results showed that clobetasol propionate and loteprednol etabonate were potent and selective CYP3A5 reversible inhibitors with selectivity of 24-fold against CYP3A4 and 39-fold or more against the other major CYPs. The selectivity of difluprednate in HLM is much weaker than that in the recombinant enzymes due to hydrolysis of the acetate group in HLM. Based on the selectivity data, loteprednol etabonate can be utilized as an orthogonal approach, when experimental fraction metabolized of CYP3A5 is greater than 0.5, to understand CYP3A5 contribution to drug metabolism and its clinical significance. Future endeavors to identify even more selective CYP3A5 inhibitors are warranted to enable accurate determination of CYP3A5 contribution to metabolism versus CYP3A4.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A , Humans , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Loteprednol Etabonate , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/metabolism
18.
Eur J Pharm Sci ; 198: 106735, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38423227

ABSTRACT

Time-dependent inhibition of cytochrome P450 (CYP) enzymes has been observed for a few glucuronide metabolites of clinically used drugs. Here, we investigated the inhibitory potential of 16 glucuronide metabolites towards nine major CYP enzymes in vitro. Automated substrate cocktail methods were used to screen time-dependent inhibition of CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2J2 and 3A in human liver microsomes. Seven glucuronides (carvedilol ß-D-glucuronide, diclofenac acyl-ß-D-glucuronide, 4-hydroxyduloxetine ß-D-glucuronide, ezetimibe phenoxy-ß-D-glucuronide, raloxifene 4'-glucuronide, repaglinide acyl-ß-D-glucuronide and valproic acid ß-D-glucuronide) caused NADPH- and time-dependent inhibition of at least one of the CYPs investigated, including CYP2A6, CYP2C19 and CYP3A. In more detailed experiments, we focused on the glucuronides of carvedilol and diclofenac, which inhibited CYP3A. Carvedilol ß-D-glucuronide showed weak time-dependent inhibition of CYP3A, but the parent drug carvedilol was found to be a more potent inhibitor of CYP3A, with the half-maximal inhibitor concentration (IC50) decreasing from 7.0 to 1.1 µM after a 30-min preincubation with NADPH. The maximal inactivation constant (kinact) and the inhibitor concentration causing half of kinact (KI) for CYP3A inactivation by carvedilol were 0.051 1/min and 1.8 µM, respectively. Diclofenac acyl-ß-D-glucuronide caused time-dependent inactivation of CYP3A at high concentrations, with a 4-fold IC50 shift (from 400 to 98 µM after a 30-min preincubation with NADPH) and KI and kinact values of >2,000 µM and >0.16 1/min. In static predictions, carvedilol caused significant (>1.25-fold) increase in the exposure of the CYP3A substrates midazolam and simvastatin. In conclusion, we identified several glucuronide metabolites with CYP inhibitory properties. Based on detailed experiments, the inactivation of CYP3A by carvedilol may cause clinically significant drug-drug interactions.


Subject(s)
Glucuronides , Microsomes, Liver , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Glucuronides/metabolism , Diclofenac/pharmacology , Diclofenac/analogs & derivatives , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism
19.
Xenobiotica ; 54(4): 195-200, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38385556

ABSTRACT

To assess the effect of Rehmannioside A on CYP450s activity and to estimate its inhibitory properties.The effect of Rehmannioside A on the activity of major CYP450s in human liver microsomes (HLMs) was assessed with the corresponding substrates and marker reactions, and compared with a blank control and the respective inhibitors. Suppression of CYP3A4, 2C9 and 2D6 was assessed by the dose-dependent assay and fitted with non-competitive or competitive inhibition models. The inhibition of CYP3A4 was determined in a time-dependent manner.Rehmannioside A suppressed the activity of CYP3A4, 2C9, and 2D6 with IC50 values of 10.08, 12.62, and 16.43 µM, respectively. Suppression of CYP3A4 was fitted to a non-competitive model with Ki value of 5.08 µM, whereas CYP2C9 and 2D6 were fitted to a competitive model with Ki values of 6.25 and 8.14 µM. Additionally, the inhibitory effect on CYP3A4 was time-dependent with KI value of 8.47 µM-1 and a Kinact of 0.048 min-1.In vitro suppression of CYP3A, 2C9 and 2D6 by Rehmannioside A indicated that Rehmannioside A or its source herbs may interact with drugs metabolised by these CYP450s, which could guide the clinical application.


Subject(s)
Cytochrome P-450 CYP3A , Microsomes, Liver , Humans , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 Enzyme Inhibitors/pharmacology
20.
CPT Pharmacometrics Syst Pharmacol ; 13(4): 589-598, 2024 04.
Article in English | MEDLINE | ID: mdl-38303579

ABSTRACT

Lefamulin is being evaluated as a treatment for bacterial exacerbations in cystic fibrosis (CF). Ivacaftor is approved for the treatment of patients with CF. Lefamulin is a moderate CYP3A inhibitor and co-administration with ivacaftor may result in a drug-drug interaction (DDI). A CF population was built based on literature using the Simcyp Simulator. A previously developed and validated physiologically-based pharmacokinetic (PBPK) model for ivacaftor was used. A PBPK model for lefamulin was developed and verified. Predicted concentrations and pharmacokinetic (PK) parameters for both ivacaftor and lefamulin in healthy subjects and patients with CF were in reasonable agreement with observed data (within 1.4-fold, majority within 1.25-fold). The lefamulin model as a CYP3A4 perpetrator was validated using a different Ki value for oral (p.o.) and intravenous (i.v.) routes. The simulated changes in area under the curve of ivacaftor in patients with CF when co-administered with p.o. and i.v. lefamulin were weak-to-moderate. The predicted change in ivacaftor PK when co-administered with oral lefamulin was less than observed between ivacaftor and fluconazole. These results suggest a low liability for a DDI between lefamulin and ivacaftor in patients with CF.


Subject(s)
Aminophenols , Cystic Fibrosis , Diterpenes , Polycyclic Compounds , Quinolones , Thioglycolates , Humans , Cystic Fibrosis/drug therapy , Quinolones/therapeutic use , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A , Drug Interactions , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...