1
|
Russell LE, Yadav J, Maldonato BJ, Chien HC, Zou L, Vergara AG, Villavicencio EG. Transporter-mediated drug-drug interactions: regulatory guidelines, in vitro and in vivo methodologies and translation, special populations, and the blood-brain barrier. Drug Metab Rev 2024:1-28. [PMID: 38967415 DOI: 10.1080/03602532.2024.2364591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/31/2024] [Indexed: 07/06/2024]
Abstract
This review, part of a special issue on drug-drug interactions (DDIs) spearheaded by the International Society for the Study of Xenobiotics (ISSX) New Investigators, explores the critical role of drug transporters in absorption, disposition, and clearance in the context of DDIs. Over the past two decades, significant advances have been made in understanding the clinical relevance of these transporters. Current knowledge on key uptake and efflux transporters that affect drug disposition and development is summarized. Regulatory guidelines from the FDA, EMA, and PMDA that inform the evaluation of potential transporter-mediated DDIs are discussed in detail. Methodologies for preclinical and clinical testing to assess potential DDIs are reviewed, with an emphasis on the utility of physiologically based pharmacokinetic (PBPK) modeling. This includes the application of relative abundance and expression factors to predict human pharmacokinetics (PK) using preclinical data, integrating the latest regulatory guidelines. Considerations for assessing transporter-mediated DDIs in special populations, including pediatric, hepatic, and renal impairment groups, are provided. Additionally, the impact of transporters at the blood-brain barrier (BBB) on the disposition of CNS-related drugs is explored. Enhancing the understanding of drug transporters and their role in drug disposition and toxicity can improve efficacy and reduce adverse effects. Continued research is essential to bridge remaining gaps in knowledge, particularly in comparison with cytochrome P450 (CYP) enzymes.
Collapse
Affiliation(s)
- Laura E Russell
- Department of Quantitative, Translational, and ADME Sciences, AbbVie Inc, North Chicago, IL, USA
| | - Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Boston, MA, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc, Redwood City, CA, USA
| | - Huan-Chieh Chien
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ling Zou
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc, South San Francisco, CA, USA
| | - Ana G Vergara
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Rahway, NJ, USA
| | - Erick G Villavicencio
- Department of Biology-Discovery, Imaging and Functional Genomics, Merck & Co., Inc, Rahway, NJ, USA
| |
Collapse
|
2
|
Hoch M, Huth F, Sato M, Sengupta T, Quinlan M, Dodd S, Kapoor S, Hourcade-Potelleret F. Pharmacokinetics of asciminib in the presence of CYP3A or P-gp inhibitors, CYP3A inducers, and acid-reducing agents. Clin Transl Sci 2022; 15:1698-1712. [PMID: 35616006 PMCID: PMC9283742 DOI: 10.1111/cts.13285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/14/2022] [Accepted: 04/06/2022] [Indexed: 12/16/2022] Open
Abstract
Asciminib is a first‐in‐class inhibitor of BCR::ABL1, specifically targeting the ABL myristoyl pocket. Asciminib is a substrate of CYP3A4 and P‐glycoprotein (P‐gp) and possesses pH‐dependent solubility in aqueous solution. This report summarizes the results of two phase I studies in healthy subjects aimed at assessing the impact of CYP3A and P‐gp inhibitors, CYP3A inducers and acid‐reducing agents (ARAs) on the pharmacokinetics (PK) of asciminib (single dose of 40 mg). Asciminib exposure (area under the curve [AUC]) unexpectedly decreased by ~40% when administered concomitantly with the strong CYP3A inhibitor itraconazole oral solution, whereas maximum plasma concentration (Cmax) decreased by ~50%. However, asciminib exposure was slightly increased in subjects receiving an itraconazole capsule (~3%) or clarithromycin (~35%), another strong CYP3A inhibitor. Macroflux studies showed that cyclodextrin (present in high quantities as excipient [40‐fold excess to itraconazole] in the oral solution formulation of itraconazole) decreased asciminib flux through a lipid membrane by ~80%. The AUC of asciminib was marginally decreased by concomitant administration with the strong CYP3A inducer rifampicin (by ~13–15%) and the strong P‐gp inhibitor quinidine (by ~13–16%). Concomitant administration of the ARA rabeprazole had little or no effect on asciminib AUC, with a 9% decrease in Cmax. The treatments were generally well tolerated. Taking into account the large therapeutic window of asciminib, the observed changes in asciminib PK following multiple doses of P‐gp, CYP3A inhibitors, CYP3A inducers, or ARAs are not considered to be clinically meaningful. Care should be exercised when administering asciminib concomitantly with cyclodextrin‐containing drug formulations.
Collapse
Affiliation(s)
- Matthias Hoch
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Felix Huth
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Masahiko Sato
- Novartis Pharma K.K., Novartis Institutes for Biomedical Research, Tokyo, Japan
| | | | | | - Stephanie Dodd
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Shruti Kapoor
- Novartis Pharmaceuticals, East Hanover, New Jersey, USA
| | | |
Collapse
|
3
|
Kohara H, Bajaj P, Yamanaka K, Miyawaki A, Harada K, Miyamoto K, Matsui T, Okai Y, Wagoner M, Shinozawa T. High-Throughput Screening to Evaluate Inhibition of Bile Acid Transporters Using Human Hepatocytes Isolated From Chimeric Mice. Toxicol Sci 2020; 173:347-361. [PMID: 31722436 DOI: 10.1093/toxsci/kfz229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cholestasis resulting from hepatic bile acid efflux transporter inhibition may contribute to drug-induced liver injury (DILI). This condition is a common safety-related reason for drug attrition and withdrawal. To screen for safety risks associated with efflux transport inhibition, we developed a high-throughput cellular assay for different drug discovery phases. Hepatocytes isolated from chimeric mice with humanized livers presented gene expression resembling that of the human liver and demonstrated apical membrane polarity when sandwiched between Matrigel and collagen. The fluorescent bile acid-derivative cholyl-l-lysyl-fluorescein (CLF) was used to quantify drug-induced efflux transport inhibition in hepatocytes. Cyclosporine inhibited CLF accumulation in the apical bile canalicular lumen in a concentration-dependent manner. The assay had equivalent predictive power to a primary human hepatocyte-based assay and greater predictive power than an assay performed with rat hepatocytes. Predictive power was tested using 45 pharmaceutical compounds, and 91.3% of the compounds with cholestatic potential (21/23) had margins (IC50/Cmax) < 20. In contrast, 90.9% (20/22) of compounds without cholestatic potential had IC50/Cmax>20. Assay sensitivity and specificity were 91.3% and 90.9%, respectively. We suggest that this improved assay performance could result from higher expression of efflux transporters, metabolic pathways, and/or species differences. Given the long-term supply of cells from the same donor, the humanized mouse-derived hepatocyte-based CLF efflux assay could be a valuable tool for predicting cholestatic DILI.
Collapse
Affiliation(s)
- Hiroshi Kohara
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| | - Piyush Bajaj
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts 02139, USA
| | - Kazunori Yamanaka
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| | - Akimitsu Miyawaki
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| | - Kosuke Harada
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| | - Kazumasa Miyamoto
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| | - Toshikatsu Matsui
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| | - Yoshiko Okai
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| | - Matthew Wagoner
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts 02139, USA
| | - Tadahiro Shinozawa
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Kanagawa, Japan
| |
Collapse
|
4
|
Tátrai P, Krajcsi P. Prediction of Drug-Induced Hyperbilirubinemia by In Vitro Testing. Pharmaceutics 2020; 12:pharmaceutics12080755. [PMID: 32796590 PMCID: PMC7465333 DOI: 10.3390/pharmaceutics12080755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022] Open
Abstract
Bilirubin, the end product of heme catabolism, is produced continuously in the body and may reach toxic levels if accumulates in the serum and tissues; therefore, a highly efficient mechanism evolved for its disposition. Normally, unconjugated bilirubin enters hepatocytes through the uptake transporters organic anion transporting polypeptide (OATP) 1B1 and 1B3, undergoes glucuronidation by the Phase II enzyme UDP glucuronosyltransferase 1A1 (UGT1A1), and conjugated forms are excreted into the bile by the canalicular export pump multidrug resistance protein 2 (MRP2). Any remaining conjugated bilirubin is transported back to the blood by MRP3 and passed on for uptake and excretion by downstream hepatocytes or the kidney. The bile salt export pump BSEP as the main motor of bile flow is indirectly involved in bilirubin disposition. Genetic mutations and xenobiotics that interfere with this machinery may impede bilirubin disposition and cause hyperbilirubinemia. Several pharmaceutical compounds are known to cause hyperbilirubinemia via inhibition of OATP1Bs, UGT1A1, or BSEP. Herein we briefly review the in vitro prediction methods that serve to identify drugs with a potential to induce hyperbilirubinemia. In vitro assays can be deployed early in drug development and may help to minimize late-stage attrition. Based on current evidence, drugs that behave as mono- or multispecific inhibitors of OATP1B1, UGT1A1, and BSEP in vitro are at risk of causing clinically significant hyperbilirubinemia. By integrating inhibition data from in vitro assays, drug serum concentrations, and clinical reports of hyperbilirubinemia, predictor cut-off values have been established and are provisionally suggested in this review. Further validation of in vitro readouts to clinical outcomes is expected to enhance the predictive power of these assays.
Collapse
Affiliation(s)
- Péter Tátrai
- Solvo Biotechnology, Science Park, Building B1, 4-20 Irinyi József utca, H-1117 Budapest, Hungary;
| | - Péter Krajcsi
- Solvo Biotechnology, Science Park, Building B1, 4-20 Irinyi József utca, H-1117 Budapest, Hungary;
- Faculty of Health Sciences, Semmelweis University, H-1085 Budapest, Hungary
- Faculty of Information Technology and Bionics, Péter Pázmány Catholic University, H-1083 Budapest, Hungary
- Correspondence:
| |
Collapse
|
5
|
Ben Salem C, Sahnoun D, Slim R, Ben Fradj F, Aouani C. Atorvastatin and sildenafil interaction-induced rhabdomyolysis. Ann Pharmacother 2020; 54:1047-1048. [PMID: 32418442 DOI: 10.1177/1060028020919933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
6
|
Peng Y, Chen L, Ye S, Kang Y, Liu J, Zeng S, Yu L. Research and development of drug delivery systems based on drug transporter and nano-formulation. Asian J Pharm Sci 2020; 15:220-236. [PMID: 32373201 PMCID: PMC7193453 DOI: 10.1016/j.ajps.2020.02.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/16/2020] [Accepted: 02/29/2020] [Indexed: 12/11/2022] Open
Abstract
In recent years, the continuous occurrence of multi-drug resistance in the clinic has made people pay more attention to the transporter. Changes in the expression and activity of transporters can cause corresponding changes in drug pharmacokinetics and pharmacodynamics. The drug-drug interactions (DDI) caused by transporters can seriously affect drug effectiveness and toxicity. In the development of pharmaceutical preparations, people have increasingly concerned about the effects and regulation of transporters in drug effects. To improve the targeting and physicochemical properties of drugs, the development of targeted agents is very rapid. Among them, novel nano-formulations are the best. With the continuous innovation and development of nano-formulation, its application has become more and more extensive. Nano-formulation has exerted certain advantages in the drug development based on transporters, and is also involved in the combination of targeted transporters. This review focuses on the application of novel nano-agents targeting transporters and the introduction of drug-transporter-based nano-formulations.
Collapse
Affiliation(s)
- Yi Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lu Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng Ye
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yu Kang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junqing Liu
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lushan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
7
|
Hougaard Christensen MM, Bruun Haastrup M, Øhlenschlaeger T, Esbech P, Arnspang Pedersen S, Bach Dunvald AC, Bjerregaard Stage T, Pilsgaard Henriksen D, Thestrup Pedersen AJ. Interaction potential between clarithromycin and individual statins-A systematic review. Basic Clin Pharmacol Toxicol 2019; 126:307-317. [PMID: 31628882 DOI: 10.1111/bcpt.13343] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022]
Abstract
The high prevalence of statin and clarithromycin utilization creates potential for overlapping use. The objectives of this MiniReview were to investigate the evidence base for drug-drug interactions between clarithromycin and currently marketed statins and to present management strategies for these drug combinations. We conducted a systematic literature review following PRISMA guidelines with English language studies retrieved from PubMed and EMBASE (from inception through March 2019). We included 29 articles (16 case reports, 5 observational, 5 clinical pharmacokinetic and 3 in vitro studies). Based on mechanistic/clinical studies involving clarithromycin or the related macrolide erythromycin (both strong inhibitors of CYP3A4 and of hepatic statin uptake transporters OATP1B1 and OATP1B3), clarithromycin is expected to substantially increase systemic exposure to simvastatin and lovastatin (>5-fold increase in area under the plasma concentration-time curve (AUC)), moderately increase AUCs of atorvastatin and pitavastatin (2- to 4-fold AUC increase) and slightly increase pravastatin exposure (≈2-fold AUC increase) while having little effect on fluvastatin or rosuvastatin. The 16 cases of statin-clarithromycin adverse drug reactions (rhabdomyolysis (n = 14) or less severe clinical myopathy) involved a CYP3A4-metabolized statin (simvastatin, lovastatin or atorvastatin). In line, a cohort study found concurrent use of clarithromycin and CYP3A4-metabolized statins to be associated with a doubled risk of hospitalization with rhabdomyolysis or other statin-related adverse events as compared with azithromycin-statin co-administration. If clarithromycin is necessary, we recommend (a) avoiding co-administration with simvastatin, lovastatin or atorvastatin; (b) withholding or dose-reducing pitavastatin; (c) continuing pravastatin therapy with caution, limiting pravastatin dose to 40 mg daily; and (d) continuing fluvastatin or rosuvastatin with caution.
Collapse
Affiliation(s)
- Mette Marie Hougaard Christensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Maija Bruun Haastrup
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Øhlenschlaeger
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Peter Esbech
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Sidsel Arnspang Pedersen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | | | - Tore Bjerregaard Stage
- Clinical Pharmacology and Pharmacy, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Daniel Pilsgaard Henriksen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark.,Clinical Pharmacology and Pharmacy, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
8
|
Shen H, Christopher L, Lai Y, Gong J, Kandoussi H, Garonzik S, Perera V, Garimella T, Humphreys WG. Further Studies to Support the Use of Coproporphyrin I and III as Novel Clinical Biomarkers for Evaluating the Potential for Organic Anion Transporting Polypeptide 1B1 and OATP1B3 Inhibition. Drug Metab Dispos 2018; 46:1075-1082. [PMID: 29777022 DOI: 10.1124/dmd.118.081125] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/16/2018] [Indexed: 12/14/2022] Open
Abstract
In a recent study, limited to South Asian Indian subjects (n = 12), coproporphyrin (CP) I and CPIII demonstrated properties appropriate for an organic anion-transporting polypeptide (OATP) 1B endogenous probe. The current studies were conducted in healthy volunteers of mixed ethnicities, including black, white, and Hispanic subjects, to better understand the utility of these biomarkers in broader populations. After oral administration with 600 mg rifampin, AUC(0-24h) values were 2.8-, 3.7-, and 3.6-fold higher than predose levels for CPI and 2.6-, 3.1-, and 2.4-fold higher for CPIII, for the three populations, respectively. These changes in response to rifampin were consistent with previous results. The sensitivity toward OATP1B inhibition was also investigated by evaluating changes of plasma CP levels in the presence of diltiazem and itraconazole [administered as part of an unrelated drug-drug interaction (DDI) investigation], two compounds that were predicted to have minimal inhibitory effect on OATP1B. Administration of diltiazem and itraconazole did not increase plasma CPI and CPIII concentrations relative to prestudy levels, in agreement with predictions from in vitro parameters. Additionally, the basal CP concentrations in subjects with SLCO1B1 c.521TT genotype were comparable to those with SLCO1B1 c.521TC genotype, similar to studies with probe substrates. However, subjects with SLCO1B1 c.388AG and c.388GG genotypes (i.e., increased OATP1B1 transport activity for certain substrates) had lower concentrations of CPI than those with SLCO1B1 c.388AA. Collectively, these findings provide further evidence supporting the translational value of CPI and CPIII as suitable endogenous clinical probes to gauge OATP1B activity and potential for OATP1B-mediated DDIs.
Collapse
Affiliation(s)
- Hong Shen
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Lisa Christopher
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Yurong Lai
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Jiachang Gong
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Hamza Kandoussi
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Samira Garonzik
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Vidya Perera
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Tushar Garimella
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| | - W Griffith Humphreys
- Metabolism and Pharmacokinetics (H.S., L.C., Y.L., J.G., W.G.H.), Bioanalytical Sciences (H.K.), and Clinical Pharmacology and Pharmacometrics (S.G., V.P., T.G.), Bristol-Myers Squibb Company, Princeton, New Jersey
| |
Collapse
|
9
|
Liu L, Cheeti S, Yoshida K, Choo E, Chen E, Chen B, Gates M, Singel S, Morley R, Ware J, Sahasranaman S. Effect of OATP1B1/1B3 Inhibitor GDC-0810 on the Pharmacokinetics of Pravastatin and Coproporphyrin I/III in Healthy Female Subjects. J Clin Pharmacol 2018; 58:1427-1435. [PMID: 29786857 DOI: 10.1002/jcph.1261] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/17/2018] [Indexed: 11/09/2022]
Abstract
Developed as an oral anticancer drug to treat estrogen receptor-positive breast cancer, GDC-0810 was shown to be a potent inhibitor of organic anion-transporting polypeptide 1B1 and 1B3 (OATP1B1/1B3) from an in vitro assay. A clinical study was conducted to assess the drug-drug interaction potential between GDC-0810 and pravastatin, which is a relatively selective and sensitive OATP1B1/1B3 substrate. Fifteen healthy female subjects of non-childbearing potential were enrolled in the study. On day 1 in period 1, a single 10-mg dose of pravastatin was administered to all subjects. Following a 4-day washout period, 600 mg of GDC-0810 was administered once daily on days 5 through 8 in period 2 to achieve steady-state concentrations. On day 7, a single dose of 10-mg pravastatin was coadministered with the 600-mg GDC-0810 dose. Concentrations of pravastatin (periods 1 and 2) and GDC-0810 (period 2 only) were quantified in blood samples and subsequently used to calculate the pharmacokinetics (PK) parameters. The pravastatin mean maximal concentration and area under the curve values were approximately 20% and 41% higher, respectively, following pravastatin coadministration with GDC-0810 compared to pravastatin alone. Based on the magnitude of change in this drug-drug interaction study, dose adjustments for pravastatin (and other OATP1B1/1B3 substrates) were not considered necessary when administered with GDC-0810. Retrospectively, the endogenous biomarkers of OATP1B1/1B3, coproporphyrin I and III, were also measured and showed changes comparable to those of pravastatin, indicating their utility in detecting weak inhibition of OATP1B1/1B3 in the clinical setting.
Collapse
Affiliation(s)
- Lichuan Liu
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Sravanthi Cheeti
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Kenta Yoshida
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Edna Choo
- Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, USA
| | - Eugene Chen
- Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, USA
| | - Buyun Chen
- Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, CA, USA
| | - Mary Gates
- Early Clinical Development, Genentech Inc., South San Francisco, CA, USA
| | - Stina Singel
- Early Clinical Development, Genentech Inc., South San Francisco, CA, USA
| | - Roland Morley
- Clinical Safety, Genentech Inc., South San Francisco, CA, USA
| | - Joseph Ware
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | | |
Collapse
|