1
|
Lou J, Li N, Jiang X, Cai X, Wang L, Wu X, Zhang W, Jin C, Zhuang X. Inhibition of P-Glycoprotein Asymmetrically Alters the In Vivo Exposure Profile of SGC003F: A Novel Guanylate Cyclase Stimulator. Pharmaceuticals (Basel) 2024; 17:1140. [PMID: 39338304 PMCID: PMC11435065 DOI: 10.3390/ph17091140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
As a novel guanylate cyclase stimulator, SGC003F is being developed for the treatment of heart failure with a reduced ejection fraction (HFrEF). This study aimed to assess the effect of P-glycoprotein (P-gp) inhibition on SGC003F exposure in vivo, comparing plasma and tissue levels, and evaluating the role of P-gp in the small intestine, blood-brain barrier (BBB), and kidney in impacting the tissue exposure. Tariquidar, a P-gp inhibitor, was added to monolayer transport assays to observe the changes in the transmembrane characteristics of SGC003F. Rats were given SGC003F with tariquidar via various routes to measure plasma, tissue, urine, and fecal concentrations. The inclusion of tariquidar significantly altered the pharmacokinetics of SGC003F. In LLC-PK1-MDR1 cells, tariquidar reduced the efflux ratio of SGC003F from 6.56 to 1.28. In rats, it enhanced the plasma AUC by 3.05 or 1.61 times, increased the Cmax by 2.13 or 1.07 times, and notably improved bioavailability from 46.4% to 95%. Additionally, co-administration with tariquidar led to a decrease in fecal excretion and an increase in tissue exposure, with only a moderate effect on the partition ratios in the small intestine and brain. P-gp inhibition impacts SGC003F exposure, with plasma levels not fully reflecting tissue levels. P-gp in the small intestine and BBB affects SGC003F's pharmacokinetics, warranting further clinical drug-drug interaction (DDI) studies.
Collapse
Affiliation(s)
- Jinle Lou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- College of Pharmay, Yanbian University, Yanji 133000, China
| | - Nan Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- College of Pharmay, Yanbian University, Yanji 133000, China
| | - Xue Jiang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xu Cai
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Lingchao Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xia Wu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wenpeng Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunmei Jin
- College of Pharmay, Yanbian University, Yanji 133000, China
| | - Xiaomei Zhuang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
2
|
Russell LE, Claw KG, Aagaard KM, Glass SM, Dasgupta K, Nez FL, Haimbaugh A, Maldonato BJ, Yadav J. Insights into pharmacogenetics, drug-gene interactions, and drug-drug-gene interactions. Drug Metab Rev 2024:1-19. [PMID: 39154360 DOI: 10.1080/03602532.2024.2385928] [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: 07/23/2024] [Indexed: 08/20/2024]
Abstract
This review explores genetic contributors to drug interactions, known as drug-gene and drug-drug-gene interactions (DGI and DDGI, respectively). This article is part of a mini-review issue led by the International Society for the Study of Xenobiotics (ISSX) New Investigators Group. Pharmacogenetics (PGx) is the study of the impact of genetic variation on pharmacokinetics (PK), pharmacodynamics (PD), and adverse drug reactions. Genetic variation in pharmacogenes, including drug metabolizing enzymes and drug transporters, is common and can increase the risk of adverse drug events or contribute to reduced efficacy. In this review, we summarize clinically actionable genetic variants, and touch on methodologies such as genotyping patient DNA to identify genetic variation in targeted genes, and deep mutational scanning as a high-throughput in vitro approach to study the impact of genetic variation on protein function and/or expression in vitro. We highlight the utility of physiologically based pharmacokinetic (PBPK) models to integrate genetic and chemical inhibitor and inducer data for more accurate human PK simulations. Additionally, we analyze the limitations of historical ethnic descriptors in pharmacogenomics research. Altogether, the work herein underscores the importance of identifying and understanding complex DGI and DDGIs with the intention to provide better treatment outcomes for patients. We also highlight current barriers to wide-scale implementation of PGx-guided dosing as standard or care in clinical settings.
Collapse
Affiliation(s)
- Laura E Russell
- Drug Metabolism and Pharmacokinetics, AbbVie Inc, North Chicago, IL, USA
| | - Katrina G Claw
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kaja M Aagaard
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah M Glass
- Preclinical Sciences and Translational Safety, Janssen Research &Development, San Diego, CA, USA
| | - Kuheli Dasgupta
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - F Leah Nez
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alex Haimbaugh
- Division of Biomedical Informatics and Personalized Medicine, CO Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin J Maldonato
- Department of Nonclinical Development and Clinical Pharmacology, Revolution Medicines, Inc, Redwood City, CA, USA
| | - Jaydeep Yadav
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc, Boston, MA, USA
| |
Collapse
|
3
|
Rask-Madsen C, Katragadda S, Li M, Ucpinar S, Chinn L, Arora P, Smith P. Effects of Quinidine or Rifampin Co-administration on the Single-Dose Pharmacokinetics and Safety of Rilzabrutinib (PRN1008) in Healthy Participants. Clin Pharmacol Drug Dev 2024; 13:590-600. [PMID: 38623935 DOI: 10.1002/cpdd.1404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/14/2024] [Indexed: 04/17/2024]
Abstract
This open-label, phase 1 study was conducted with healthy adult participants to evaluate the potential drug-drug interaction between rilzabrutinib and quinidine (an inhibitor of P-glycoprotein [P-gp] and CYP2D6) or rifampin (an inducer of CYP3A and P-gp). Plasma concentrations of rilzabrutinib were measured after a single oral dose of rilzabrutinib 400 mg administered on day 1 and again, following a wash-out period, after co-administration of rilzabrutinib and quinidine or rifampin. Specifically, quinidine was given at a dose of 300 mg every 8 hours for 5 days from day 7 to day 11 (N = 16) while rifampin was given as 600 mg once daily for 11 days from day 7 to day 17 (N = 16) with rilzabrutinib given in the morning of day 10 (during quinidine dosing) or day 16 (during rifampin dosing). Quinidine had no significant effect on rilzabrutinib pharmacokinetics. Rifampin decreased rilzabrutinib exposure (the geometric mean of Cmax and AUC0-∞ decreased by 80.5% and 79.5%, respectively). Single oral doses of rilzabrutinib, with or without quinidine or rifampin, appeared to be well tolerated. These findings indicate that rilzabrutinib is a substrate for CYP3A but not a substrate for P-gp.
Collapse
Affiliation(s)
| | - Suresh Katragadda
- Department of Pharmacokinetics, Dynamics and Metabolism, Sanofi, Cambridge, MA, USA
| | - Mengyao Li
- Department of Pharmacokinetics, Dynamics and Metabolism, Sanofi, Bridgewater, NJ, USA
| | - Sibel Ucpinar
- Department of Pharmacokinetics, Dynamics and Metabolism, Sanofi, Bridgewater, NJ, USA
| | - Leslie Chinn
- Department of Pharmacokinetics, Dynamics and Metabolism, Sanofi, Bridgewater, NJ, USA
| | - Puneet Arora
- Department of Clinical, Inflammation and Immunology, Sanofi, South San Francisco, CA, USA
| | - Patrick Smith
- Integrated Drug Development, Certara, Parsippany, NJ, USA
| |
Collapse
|
4
|
Peruzzi E, Roncato R, De Mattia E, Bignucolo A, Swen JJ, Guchelaar HJ, Toffoli G, Cecchin E. Implementation of pre-emptive testing of a pharmacogenomic panel in clinical practice: Where do we stand? Br J Clin Pharmacol 2023. [PMID: 37926674 DOI: 10.1111/bcp.15956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
Adverse drug reactions (ADRs) account for a large proportion of hospitalizations among adults and are more common in multimorbid patients, worsening clinical outcomes and burdening healthcare resources. Over the past decade, pharmacogenomics has been developed as a practical tool for optimizing treatment outcomes by mitigating the risk of ADRs. Some single-gene reactive tests are already used in clinical practice, including the DPYD test for fluoropyrimidines, which demonstrates how integrating pharmacogenomic data into routine care can improve patient safety in a cost-effective manner. The evolution from reactive single-gene testing to comprehensive pre-emptive genotyping panels holds great potential for refining drug prescribing practices. Several implementation projects have been conducted to test the feasibility of applying different genetic panels in clinical practice. Recently, the results of a large prospective randomized trial in Europe (the PREPARE study by Ubiquitous Pharmacogenomics consortium) have provided the first evidence that prospective application of a pre-emptive pharmacogenomic test panel in clinical practice, in seven European healthcare systems, is feasible and yielded a 30% reduction in the risk of developing clinically relevant toxicities. Nevertheless, some important questions remain unanswered and will hopefully be addressed by future dedicated studies. These issues include the cost-effectiveness of applying a pre-emptive genotyping panel, the role of multiple co-medications, the transferability of currently tested pharmacogenetic guidelines among patients of non-European origin and the impact of rare pharmacogenetic variants that are not detected by currently used genotyping approaches.
Collapse
Affiliation(s)
- Elena Peruzzi
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Rossana Roncato
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
- Department of Medicine, University of Udine, Udine, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Alessia Bignucolo
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano, Istituti di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| |
Collapse
|