1
|
Li J, Parsley E, Cravets M, DeNoia E, Key C, Mathias A. Phase 1 Studies to Assess Inhaled Seralutinib as a Perpetrator or a Victim of Drug-Drug Interactions in Healthy Participants. Clin Pharmacol Drug Dev 2025; 14:91-104. [PMID: 39711098 DOI: 10.1002/cpdd.1491] [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: 06/25/2024] [Accepted: 11/11/2024] [Indexed: 12/24/2024]
Abstract
Seralutinib, an inhaled, small-molecule tyrosine kinase inhibitor in clinical development for the treatment of pulmonary arterial hypertension (PAH), was evaluated for its potential as a perpetrator or victim of a metabolic and transporter-based drug-drug interactions in 2 phase 1 studies. In study 1, 24 participants received a cocktail of probe substrates: caffeine (CYP1A2), montelukast (CYP2C8), flurbiprofen (CYP2C9), midazolam (CYP3A), and pravastatin (OATP1B1/1B3), plus digoxin (P-gp) with or without seralutinib. In study 2, 19 participants received seralutinib with/without itraconazole, a strong CYP3A inhibitor, or fosaprepitant, a weak CYP3A inhibitor. Geometric least-squares mean ratios and 90% confidence intervals for maximum observed concentration (Cmax) and area under the plasma concentration-time curve (AUC) were obtained. Safety was monitored throughout the studies. All adverse events were mild or moderate in severity. Seralutinib coadministration increased AUC for midazolam 3.03-fold and caffeine 1.32-fold. The coadministration increased digoxin Cmax 1.28-fold. Seralutinib did not meaningfully alter Cmax and AUC for montelukast, flurbiprofen, or pravastatin. Fosaprepitant and itraconazole increased seralutinib AUC 1.08- and 1.84-fold, respectively. Seralutinib is a moderate CYP3A inhibitor and a weak CYP1A2 inhibitor; it slightly inhibits P-gp. Seralutinib exposure is minimally affected by a weak CYP3A inhibitor but is substantially increased by a strong CYP3A inhibitor.
Collapse
Affiliation(s)
- Jianke Li
- Gossamer Bio, Inc., San Diego, CA, USA
| | | | | | | | | | | |
Collapse
|
2
|
Rahm AK, Hackbarth J, Müller ME, Pfeiffer J, Gampp H, Petersenn F, Rivinius R, Frey N, Lugenbiel P, Thomas D. Differential Effects of the Betablockers Carvedilol, Metoprolol and Bisoprolol on Cardiac K v4.3 (I to) Channel Isoforms. Int J Mol Sci 2023; 24:13842. [PMID: 37762145 PMCID: PMC10530285 DOI: 10.3390/ijms241813842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiac Kv4.3 channels contribute to the transient outward K+ current, Ito, during early repolarization of the cardiac action potential. Two different isoforms of Kv4.3 are present in the human ventricle and exhibit differential remodeling in heart failure (HF). Cardioselective betablockers are a cornerstone of HF with reduced ejection fraction therapy as well as ventricular arrhythmia treatment. In this study we examined pharmacological effects of betablockers on both Kv4.3 isoforms to explore their potential for isoform-specific therapy. Kv4.3 isoforms were expressed in Xenopus laevis oocytes and incubated with the respective betablockers. Dose-dependency and biophysical characteristics were examined. HEK 293T-cells were transfected with the two Kv4.3 isoforms and analyzed with Western blots. Carvedilol (100 µM) blocked Kv4.3 L by 77 ± 2% and Kv4.3 S by 67 ± 6%, respectively. Metoprolol (100 µM) was less effective with inhibition of 37 ± 3% (Kv4.3 L) and 35 ± 4% (Kv4.3 S). Bisoprolol showed no inhibitory effect. Current reduction was not caused by changes in Kv4.3 protein expression. Carvedilol inhibited Kv4.3 channels at physiologically relevant concentrations, affecting both isoforms. Metoprolol showed a weaker blocking effect and bisoprolol did not exert an effect on Kv4.3. Blockade of repolarizing Kv4.3 channels by carvedilol and metoprolol extend their pharmacological mechanism of action, potentially contributing beneficial antiarrhythmic effects in normal and failing hearts.
Collapse
Affiliation(s)
- Ann-Kathrin Rahm
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Juline Hackbarth
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Mara E. Müller
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Julia Pfeiffer
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Heike Gampp
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Finn Petersenn
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Rasmus Rivinius
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Norbert Frey
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Patrick Lugenbiel
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Dierk Thomas
- Heidelberg Center for Heart Rhythm Disorders, Heidelberg University Hospital, 69120 Heidelberg, Germany (M.E.M.); (R.R.); (P.L.)
- Department of Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
3
|
Carucci M, Duez J, Tarning J, García-Barbazán I, Fricot-Monsinjon A, Sissoko A, Dumas L, Gamallo P, Beher B, Amireault P, Dussiot M, Dao M, Hull MV, McNamara CW, Roussel C, Ndour PA, Sanz LM, Gamo FJ, Buffet P. Safe drugs with high potential to block malaria transmission revealed by a spleen-mimetic screening. Nat Commun 2023; 14:1951. [PMID: 37029122 PMCID: PMC10082216 DOI: 10.1038/s41467-023-37359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/15/2023] [Indexed: 04/09/2023] Open
Abstract
Malaria parasites like Plasmodium falciparum multiply in red blood cells (RBC), which are cleared from the bloodstream by the spleen when their deformability is altered. Drug-induced stiffening of Plasmodium falciparum-infected RBC should therefore induce their elimination from the bloodstream. Here, based on this original mechanical approach, we identify safe drugs with strong potential to block the malaria transmission. By screening 13 555 compounds with spleen-mimetic microfilters, we identified 82 that target circulating transmissible form of P. falciparum. NITD609, an orally administered PfATPase inhibitor with known effects on P. falciparum, killed and stiffened transmission stages in vitro at nanomolar concentrations. Short exposures to TD-6450, an orally-administered NS5A hepatitis C virus inhibitor, stiffened transmission parasite stages and killed asexual stages in vitro at high nanomolar concentrations. A Phase 1 study in humans with a primary safety outcome and a secondary pharmacokinetics outcome ( https://clinicaltrials.gov , ID: NCT02022306) showed no severe adverse events either with single or multiple doses. Pharmacokinetic modelling showed that these concentrations can be reached in the plasma of subjects receiving short courses of TD-6450. This physiologically relevant screen identified multiple mechanisms of action, and safe drugs with strong potential as malaria transmission-blocking agents which could be rapidly tested in clinical trials.
Collapse
Affiliation(s)
- Mario Carucci
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | | | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 10400, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Irene García-Barbazán
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, 28222, Madrid, Spain
| | - Aurélie Fricot-Monsinjon
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Abdoulaye Sissoko
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Lucie Dumas
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Pablo Gamallo
- Global Health Medicines R&D, GlaxoSmith Kline (GSK), 28760, Tres Cantos, Spain
| | - Babette Beher
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Pascal Amireault
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
- Laboratory of cellular and molecular mechanisms of hematological disorders and therapeutic implications, INSERM, 75014, Paris, France
| | - Michael Dussiot
- Laboratory of cellular and molecular mechanisms of hematological disorders and therapeutic implications, INSERM, 75014, Paris, France
- Laboratoire d'Excellence GR-Ex, Paris, France
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, MA, 02139, Cambridge, USA
| | - Mitchell V Hull
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Case W McNamara
- Calibr, a division of The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Camille Roussel
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
- Laboratoire d'Excellence GR-Ex, Paris, France
- Laboratoire d'Hématologie générale, Hôpital Universitaire Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015, Paris, France
| | - Papa Alioune Ndour
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France
| | - Laura Maria Sanz
- Global Health Medicines R&D, GlaxoSmith Kline (GSK), 28760, Tres Cantos, Spain
| | | | - Pierre Buffet
- Université Paris Cité, Inserm, UMR-1134, Biologie Intégré du Globule Rouge, 75015, Paris, France.
- Department of Infectious & Tropical Disease, AP-HP, Necker Hospital, 75015, Paris, France.
- Centre Médical de l'Institut Pasteur (CMIP), Institut Pasteur, 75015, Paris, France.
| |
Collapse
|
4
|
Pozzi C, Vanet A, Francesconi V, Tagliazucchi L, Tassone G, Venturelli A, Spyrakis F, Mazzorana M, Costi MP, Tonelli M. Antitarget, Anti-SARS-CoV-2 Leads, Drugs, and the Drug Discovery-Genetics Alliance Perspective. J Med Chem 2023; 66:3664-3702. [PMID: 36857133 PMCID: PMC10005815 DOI: 10.1021/acs.jmedchem.2c01229] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The most advanced antiviral molecules addressing major SARS-CoV-2 targets (Main protease, Spike protein, and RNA polymerase), compared with proteins of other human pathogenic coronaviruses, may have a short-lasting clinical efficacy. Accumulating knowledge on the mechanisms underlying the target structural basis, its mutational progression, and the related biological significance to virus replication allows envisaging the development of better-targeted therapies in the context of COVID-19 epidemic and future coronavirus outbreaks. The identification of evolutionary patterns based solely on sequence information analysis for those targets can provide meaningful insights into the molecular basis of host-pathogen interactions and adaptation, leading to drug resistance phenomena. Herein, we will explore how the study of observed and predicted mutations may offer valuable suggestions for the application of the so-called "synthetic lethal" strategy to SARS-CoV-2 Main protease and Spike protein. The synergy between genetics evidence and drug discovery may prioritize the development of novel long-lasting antiviral agents.
Collapse
Affiliation(s)
- Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy,
University of Siena, via Aldo Moro 2, 53100 Siena,
Italy
| | - Anne Vanet
- Université Paris Cité,
CNRS, Institut Jacques Monod, F-75013 Paris,
France
| | - Valeria Francesconi
- Department of Pharmacy, University of
Genoa, viale Benedetto XV n.3, 16132 Genoa, Italy
| | - Lorenzo Tagliazucchi
- Department of Life Science, University of
Modena and Reggio Emilia, via Campi 103, 41125 Modena,
Italy
- Doctorate School in Clinical and Experimental Medicine
(CEM), University of Modena and Reggio Emilia, Via Campi 287,
41125 Modena, Italy
| | - Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy,
University of Siena, via Aldo Moro 2, 53100 Siena,
Italy
| | - Alberto Venturelli
- Department of Life Science, University of
Modena and Reggio Emilia, via Campi 103, 41125 Modena,
Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology,
University of Turin, Via Giuria 9, 10125 Turin,
Italy
| | - Marco Mazzorana
- Diamond Light Source, Harwell Science and
Innovation Campus, Didcot, Oxfordshire OX11 0DE,
U.K.
| | - Maria P. Costi
- Department of Life Science, University of
Modena and Reggio Emilia, via Campi 103, 41125 Modena,
Italy
| | - Michele Tonelli
- Department of Pharmacy, University of
Genoa, viale Benedetto XV n.3, 16132 Genoa, Italy
| |
Collapse
|
5
|
Barth A, Perry CR, Shabbir S, Zamek-Gliszczynski MJ, Thomas S, Dumont EF, Brimhall DB, Nguyen D, Srinivasan M, Swift B. Clinical assessment of gepotidacin (GSK2140944) as a victim and perpetrator of drug-drug interactions via CYP3A metabolism and transporters. Clin Transl Sci 2023; 16:647-661. [PMID: 36642822 PMCID: PMC10087077 DOI: 10.1111/cts.13477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/14/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023] Open
Abstract
Gepotidacin is a novel triazaacenaphthylene antibiotic in phase III development. Based on nonclinical in vitro characterization of gepotidacin metabolism, two phase I studies were conducted in healthy participants to investigate clinical drug-drug interactions (DDIs). We assessed gepotidacin as a DDI victim with a potent cytochrome P450 (CYP) 3A4/P-glycoprotein (P-gp) inhibitor (itraconazole), potent CYP3A4 inducer (rifampicin), and nonspecific organic cation transporter (OCT)/multidrug and toxic extrusion transporter (MATE) renal transport inhibitor (cimetidine) via single doses of gepotidacin before and after co-administration with multiple doses of the modulator drugs. Gepotidacin DDI perpetrator potential for P-gp inhibition (digoxin) and CYP3A4 inhibition (midazolam) was evaluated via single doses of the two-drug cocktail without and with gepotidacin. The DDI magnitudes were interpreted based on area under the concentration-time curve (AUC). A weak DDI (AUC increase 48%-50%) was observed for gepotidacin co-administered with itraconazole. A clinically significant decrease in gepotidacin plasma AUC (52%) was observed with rifampicin coadministration, indicating a moderate DDI. There was no DDI for gepotidacin with cimetidine; a unique biomarker approach showed increased serum creatinine (24%), decreased renal clearance of creatinine (21%), and N1-methylnicotinamide (39%), which confirmed extensive MATE inhibition and partial OCT2 inhibition. Gepotidacin was not a P-gp DDI perpetrator, although the maximum plasma concentration of digoxin increased (53%) and is potentially clinically relevant given its narrow therapeutic index. Gepotidacin demonstrated weak CYP3A4 inhibition with midazolam (<2-fold AUC increase). There were no new safety-risk profile findings. These results will inform the safe and efficacious clinical use of gepotidacin when co-administered with other drugs.
Collapse
Affiliation(s)
- Aline Barth
- Global Blood Therapeutics, South San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Kamath A, Srinivasamurthy SK, Chowta MN, Ullal SD, Daali Y, Chakradhara Rao US. Role of Drug Transporters in Elucidating Inter-Individual Variability in Pediatric Chemotherapy-Related Toxicities and Response. Pharmaceuticals (Basel) 2022; 15:990. [PMID: 36015138 PMCID: PMC9415926 DOI: 10.3390/ph15080990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Pediatric cancer treatment has evolved significantly in recent decades. The implementation of risk stratification strategies and the selection of evidence-based chemotherapy combinations have improved survival outcomes. However, there is large interindividual variability in terms of chemotherapy-related toxicities and, sometimes, the response among this population. This variability is partly attributed to the functional variability of drug-metabolizing enzymes (DME) and drug transporters (DTS) involved in the process of absorption, distribution, metabolism and excretion (ADME). The DTS, being ubiquitous, affects drug disposition across membranes and has relevance in determining chemotherapy response in pediatric cancer patients. Among the factors affecting DTS function, ontogeny or maturation is important in the pediatric population. In this narrative review, we describe the role of drug uptake/efflux transporters in defining pediatric chemotherapy-treatment-related toxicities and responses. Developmental differences in DTS and the consequent implications are also briefly discussed for the most commonly used chemotherapeutic drugs in the pediatric population.
Collapse
Affiliation(s)
- Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Mukta N. Chowta
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Sheetal D. Ullal
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Youssef Daali
- Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Uppugunduri S. Chakradhara Rao
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
7
|
Hua Y, Dai X, Xu Y, Xing G, Liu H, Lu T, Chen Y, Zhang Y. Drug repositioning: Progress and challenges in drug discovery for various diseases. Eur J Med Chem 2022; 234:114239. [PMID: 35290843 PMCID: PMC8883737 DOI: 10.1016/j.ejmech.2022.114239] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 02/20/2022] [Accepted: 02/24/2022] [Indexed: 12/17/2022]
Abstract
Compared with traditional de novo drug discovery, drug repurposing has become an attractive drug discovery strategy due to its low-cost and high efficiency. Through a comprehensive analysis of the candidates that have been identified with drug repositioning potentials, it is found that although some drugs do not show obvious advantages in the original indications, they may exert more obvious effects in other diseases. In addition, some drugs have a synergistic effect to exert better clinical efficacy if used in combination. Particularly, it has been confirmed that drug repositioning has benefits and values on the current public health emergency such as the COVID-19 pandemic, which proved the great potential of drug repositioning. In this review, we systematically reviewed a series of representative drugs that have been repositioned for different diseases and illustrated successful cases in each disease. Especially, the mechanism of action for the representative drugs in new indications were explicitly explored for each disease, we hope this review can provide important insights for follow-up research.
Collapse
Affiliation(s)
- Yi Hua
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Xiaowen Dai
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Yuan Xu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Guomeng Xing
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Haichun Liu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Tao Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| | - Yanmin Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
8
|
Chu X, Chan GH, Houle R, Lin M, Yabut J, Fandozzi C. In Vitro Assessment of Transporter Mediated Perpetrator DDIs for Several Hepatitis C Virus Direct-Acting Antiviral Drugs and Prediction of DDIs with Statins Using Static Models. AAPS J 2022; 24:45. [DOI: 10.1208/s12248-021-00677-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/21/2021] [Indexed: 01/04/2023] Open
|
9
|
Huličiak M, Vokřál I, Holas O, Martinec O, Štaud F, Červený L. Evaluation of the Potency of Anti-HIV and Anti-HCV Drugs to Inhibit P-Glycoprotein Mediated Efflux of Digoxin in Caco-2 Cell Line and Human Precision-Cut Intestinal Slices. Pharmaceuticals (Basel) 2022; 15:ph15020242. [PMID: 35215354 PMCID: PMC8875242 DOI: 10.3390/ph15020242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/23/2022] Open
Abstract
The inhibition of P-glycoprotein (ABCB1) could lead to increased drug plasma concentrations and hence increase drug toxicity. The evaluation of a drug’s ability to inhibit ABCB1 is complicated by the presence of several transport-competent sites within the ABCB1 binding pocket, making it difficult to select appropriate substrates. Here, we investigate the capacity of antiretrovirals and direct-acting antivirals to inhibit the ABCB1-mediated intestinal efflux of [3H]-digoxin and compare it with our previous rhodamine123 study. At concentrations of up to 100 µM, asunaprevir, atazanavir, daclatasvir, darunavir, elbasvir, etravirine, grazoprevir, ledipasvir, lopinavir, rilpivirine, ritonavir, saquinavir, and velpatasvir inhibited [3H]-digoxin transport in Caco-2 cells and/or in precision-cut intestinal slices prepared from the human jejunum (hPCIS). However, abacavir, dolutegravir, maraviroc, sofosbuvir, tenofovir disoproxil fumarate, and zidovudine had no inhibitory effect. We thus found that most of the tested antivirals have a high potential to cause drug–drug interactions on intestinal ABCB1. Comparing the Caco-2 and hPCIS experimental models, we conclude that the Caco-2 transport assay is more sensitive, but the results obtained using hPCIS agree better with reported in vivo observations. More inhibitors were identified when using digoxin as the ABCB1 probe substrate than when using rhodamine123. However, both approaches had limitations, indicating that inhibitory potency should be tested with at least these two ABCB1 probes.
Collapse
Affiliation(s)
- Martin Huličiak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic; (M.H.); (O.M.); (F.Š.); (L.Č.)
| | - Ivan Vokřál
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic; (M.H.); (O.M.); (F.Š.); (L.Č.)
- Correspondence:
| | - Ondřej Holas
- Department of Pharmaceutical Technology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic;
| | - Ondřej Martinec
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic; (M.H.); (O.M.); (F.Š.); (L.Č.)
| | - František Štaud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic; (M.H.); (O.M.); (F.Š.); (L.Č.)
| | - Lukáš Červený
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic; (M.H.); (O.M.); (F.Š.); (L.Č.)
| |
Collapse
|
10
|
Talluri S. Molecular Docking and Virtual Screening Based Prediction of Drugs for COVID-19. Comb Chem High Throughput Screen 2021; 24:716-728. [PMID: 32798373 DOI: 10.2174/1386207323666200814132149] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/08/2020] [Accepted: 07/14/2020] [Indexed: 11/22/2022]
Abstract
AIMS To predict potential drugs for COVID-19 by using molecular docking for virtual screening of drugs approved for other clinical applications. BACKGROUND SARS-CoV-2 is the betacoronavirus responsible for the COVID-19 pandemic. It was listed as a potential global health threat by the WHO due to high mortality, high basic reproduction number, and lack of clinically approved drugs and vaccines. The genome of the virus responsible for COVID-19 has been sequenced. In addition, the three-dimensional structure of the main protease has been determined experimentally. OBJECTIVE To identify potential drugs that can be repurposed for treatment of COVID-19 by using molecular docking based virtual screening of all approved drugs. METHODS A list of drugs approved for clinical use was obtained from the SuperDRUG2 database. The structure of the target in the apo form, as well as structures of several target-ligand complexes, were obtained from RCSB PDB. The structure of SARS-CoV-2 Mpro determined from X-ray diffraction data was used as the target. Data regarding drugs in clinical trials for COVID-19 was obtained from clinicaltrials.org. Input for molecular docking based virtual screening was prepared by using Obabel and customized python, bash, and awk scripts. Molecular docking calculations were carried out with Vina and SMINA, and the docked conformations were analyzed and visualized with PLIP, Pymol, and Rasmol. RESULTS Among the drugs that are being tested in clinical trials for COVID-19, Danoprevir and Darunavir were predicted to have the highest binding affinity for the Main protease (Mpro) target of SARS-CoV-2. Saquinavir and Beclabuvir were identified as the best novel candidates for COVID-19 therapy by using Virtual Screening of drugs approved for other clinical indications. CONCLUSION Protease inhibitors approved for treatment of other viral diseases have the potential to be repurposed for treatment of COVID-19.
Collapse
Affiliation(s)
- Sekhar Talluri
- Department of Biotechnology, GITAM, Visakhapatnam, India
| |
Collapse
|
11
|
El-Kimary EI, Ragab MAA. Recent Analytical Methodologies for the Determination of Omeprazole and/or Its Active Isomer Esomeprazole in Different Matrices: A Critical Review. Crit Rev Anal Chem 2020; 52:106-130. [DOI: 10.1080/10408347.2020.1791042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Eman I. El-Kimary
- Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, University of Alexandria, El-Messalah, Alexandria, Egypt
| | - Marwa A. A. Ragab
- Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, University of Alexandria, El-Messalah, Alexandria, Egypt
| |
Collapse
|
12
|
Smolders EJ, Ter Horst PJG, Wolters S, Burger DM. Cardiovascular Risk Management and Hepatitis C: Combining Drugs. Clin Pharmacokinet 2020; 58:565-592. [PMID: 30259390 PMCID: PMC6451722 DOI: 10.1007/s40262-018-0710-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Direct-acting antivirals (DAAs) are known victims (substrate) and perpetrators (cause) of drug–drug interactions (DDIs). These DAAs are used for the treatment of hepatitis C virus (HCV) infections and are highly effective drugs. Drugs used for cardiovascular risk management are frequently used by HCV-infected patients, whom also are treated with DAAs. Therefore, the aim of this review was to describe DDIs between cardiovascular drugs (CVDs) and DAAs. An extensive literature search was performed containing search terms for the marketed DAAs and CVDs (β-blocking agents, ACE inhibitors, angiotensin II antagonists, renin inhibitors, diuretics, calcium channel blockers, statins/ezetimibe, fibrates, platelet aggregation inhibitors, vitamin K antagonists, heparins, direct Xa inhibitors, nitrates, amiodarone, and digoxin). In particular, the drug labels from the European Medicines Agency and the US Food and Drug Administration were used. A main finding of this review is that CVDs are mostly victims of DDIs with DAAs. Therefore, when possible, monitoring of pharmacodynamics is recommended when coadministering these drugs with DAAs. Nevertheless, it is sometimes better to discontinue a drug on a temporary basis (statins, ezetimide). The DAAs are victims of DDIs in combination with bisoprolol, carvedilol, labetalol, verapamil, and gemfibrozil. Despite there are many DDIs predicted in this review, most of these DDIs can be managed by monitoring the efficacy and toxicity of the victim drug or by switching to another CVD/DAA.
Collapse
Affiliation(s)
- Elise J Smolders
- Department of Pharmacy, Isala Hospital, Dokter van Heesweg 2, 8025 AB, Zwolle, The Netherlands. .,Department of Pharmacy, Radboud university medical center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
| | - Peter J G Ter Horst
- Department of Pharmacy, Isala Hospital, Dokter van Heesweg 2, 8025 AB, Zwolle, The Netherlands
| | - Sharon Wolters
- Department of Pharmacy, Isala Hospital, Dokter van Heesweg 2, 8025 AB, Zwolle, The Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud university medical center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Zappulo E, Scotto R, Buonomo AR, Maraolo AE, Pinchera B, Gentile I. Efficacy and safety of a fixed dose combination tablet of asunaprevir + beclabuvir + daclatasvir for the treatment of Hepatitis C. Expert Opin Pharmacother 2020; 21:261-273. [PMID: 31914336 DOI: 10.1080/14656566.2019.1697674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Hepatitis C virus (HCV) is estimated to infect approximately 70 million people worldwide. If left untreated, chronic infection can progress to cirrhosis, liver failure or hepatocellular carcinoma. The advent of new direct-acting antivirals (DAA) has revolutionized patients' chances of treatment and viral elimination. Currently, several DAA options are available on the market.Areas covered: This review focuses on the pharmacokinetics, efficacy, tolerability and safety profile of DCV-TRIO, a twice-daily fixed-dose combination of daclatasvir, asunaprevir and beclabuvir approved in Japan for the treatment of genotype 1 HCV infection.Expert opinion: The DCV-TRIO combination achieved good response rates in genotype 1 patients (SVR12 ≥ 95% in naïve subtype 1b), independently from IL28B genotype, cirrhotic status and prior interferon exposure. On the other hand, unsatisfying response rates were reported in DAA-experienced patients and the risk of RAS selection should not be underestimated. Moreover, DCV-TRIO lacks differentiation from its earlier-launched DAA rivals, presents an inconvenient twice-daily dosing schedule and is not recommended in patients with advanced liver and kidney disease. All these drawbacks considerably limit its effective commercial potential. However, it can be a therapeutic option against HCV in tailored approaches according to the needs of different markets across the world.Abbreviations AE: adverse event; ALT: alanine aminotransferase; AST: aspartate aminotransferase; ASV: asunaprevir; AUC: area under the curve; BCRP: Breast Cancer Resistance Protein; BCV: boceprevir; BID: bis in die; CI: confidence intervals; CLcr: creatinine clearance; DAA: direct acting antivirals; DCV: daclatasvir; EC50: Half maximal effective concentration; GT: genotype; HCV: Hepatitis C virus; IFN: Interferon; NHL: non-Hodgkin lymphoma; OATP: Organic anion transporting polypeptides; OR: odds ratio; P-gp: P-glycoprotein; PK: pharmacokinetics; QD: quo die; RAS: resistance-associated substitutions; SVR: sustained virological response; USD: Unites States dollar.
Collapse
Affiliation(s)
- Emanuela Zappulo
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| | - Riccardo Scotto
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| | - Antonio Riccardo Buonomo
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| | - Alberto Enrico Maraolo
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| | - Biagio Pinchera
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| | - Ivan Gentile
- Department of Clinical Medicine and Surgery, Section of Infectious Diseases, University of Naples Federico II, Naples, Italy
| |
Collapse
|
14
|
Giri P, Patel H, Srinivas NR. Use of Cocktail Probe Drugs for Indexing Cytochrome P450 Enzymes in Clinical Pharmacology Studies - Review of Case Studies. Drug Metab Lett 2020; 13:3-18. [PMID: 30451124 DOI: 10.2174/1872312812666181119154734] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND The cocktail approach of probing drug metabolizing enzymes, in particular cytochrome P450 (CYP) enzymes, is a cornerstone in clinical pharmacology studies. The first report of the famous "Pittsburg cocktail" has led the way for the availability of numerous cocktail substrate mixtures that provide options for indexing of CYP enzymes and/or evaluating the perpetrator capacity of the drug. OBJECTIVE The key objectives were: 1) To collate, tabulate, and discuss the various cocktail substrates to determine specific CYP enzyme activity in clinical pharmacology studies with specific case studies; 2) To introspect on how the cocktail approach has withstood the test of time and evolved for enabling key decision(s); 3) To provide some futuristic views on the use of cocktail in drug discovery and development. METHOD The review was compiled after consultation with databases such as PubMed (NCBI database) and Google scholar to source various published literature on cocktail approaches in drug development. RESULTS In the reviewed case studies, CYP indexing was achieved using a single time point (differing for specific CYP enzyme) plasma determination of the metabolite to parent ratio for all CYP enzymes with the exception of CYP3A4/5, where multiple time points were required for exposure measurement of midazolam and its metabolite. Likewise, a single void of urine, for a specific time duration, has been utilized for the recovery measurements of parent and metabolite for CYP indexing purposes. CONCLUSION The review provides a comprehensive list of various types of cocktail approaches and discusses some key considerations including the evolution of the cocktail approaches over time, perspectives and futuristic views for the use of probe drugs to aid the execution of clinical pharmacology studies and data interpretation.
Collapse
Affiliation(s)
- Poonam Giri
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Sarkhej-Bavla N.H. No. 8A, Moraiya. Tal: Sanand, Ahmedabad-382 210, India
| | - Harilal Patel
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Sarkhej-Bavla N.H. No. 8A, Moraiya. Tal: Sanand, Ahmedabad-382 210, India
| | - Nuggehally R Srinivas
- Department of Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Sarkhej-Bavla N.H. No. 8A, Moraiya. Tal: Sanand, Ahmedabad-382 210, India.,Suramus Bio, Drug Development, J.P. Nagar First Phase, Bangalore 560078, India
| |
Collapse
|
15
|
Recent progress in in vivo phenotyping technologies for better prediction of transporter-mediated drug-drug interactions. Drug Metab Pharmacokinet 2020; 35:76-88. [PMID: 31948854 DOI: 10.1016/j.dmpk.2019.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 12/20/2022]
Abstract
Clinical reports on transporter-mediated drug-drug interactions (TP-DDIs) have rapidly accumulated and regulatory guidance/guidelines recommend that sponsors consider performing quantitative prediction of TP-DDI risks in the process of drug development. In vitro experiments for characterizing the function of drug transporters have been established and various parameters such as the inhibition constant (Ki) of drugs and the intrinsic uptake/efflux clearance for a certain transporter can be obtained. However, many reports have indicated large discrepancies between the parameters estimated from in vitro experiments and those rationally explaining drug pharmacokinetics. Thus, it is essential to evaluate directly the function of each transporter isoform in vivo in humans. At present, several transporter substrate drugs and endogenous compounds have been recognized as probe substrates for a specific transporter and transporter function was evaluated by monitoring the plasma and urine concentration of those probes; however, few compounds specifically transported via a single transporter isoform have been found. For monitoring the intraorgan concentration of drugs, positron emission tomography can be a powerful tool and clinical examples for quantification of in vivo transporter function have been published. In this review, novel methodologies for in vivo phenotyping of transporter function are summarized.
Collapse
|
16
|
Matsumoto J, San SN, Fujiyoshi M, Kawauchi A, Chiba N, Tagai R, Sanbe R, Yanaka S, Sakaue H, Kato Y, Nakamura H, Yamada H, Ariyoshi N. Effect of CYP3A5*3 genetic variant on the metabolism of direct-acting antivirals in vitro: a different effect on asunaprevir versus daclatasvir and beclabuvir. J Hum Genet 2019; 65:143-153. [PMID: 31645655 DOI: 10.1038/s10038-019-0685-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/29/2019] [Accepted: 10/02/2019] [Indexed: 12/18/2022]
Abstract
Direct-acting antivirals, asunaprevir (ASV), daclatasvir (DCV), and beclabuvir (BCV) are known to be mainly metabolized by CYP3A enzymes; however, the differences in the detailed metabolic activities of CYP3A4 and CYP3A5 on these drugs are not well clarified. The aim of the present study was to elucidate the relative contributions of CYP3A4 and CYP3A5 to the metabolism of ASV, DCV, and BCV, as well as the effect of CYP3A5*3 genetic variant in vitro. The amount of each drug and their major metabolites were determined using LC-MS/MS. Recombinant CYP3As and CYP3A5*3-genotyped human liver microsomes (CYP3A5 expressers or non-expressers) were used for the determination of their metabolic activities. The contribution of CYP3A5 to ASV metabolism was considerable compared to that of CYP3A4. Consistently, ASV metabolic activity in CYP3A5 expressers was higher than those in CYP3A5 non-expresser. Moreover, CYP3A5 expression level was significantly correlated with ASV metabolism. In contrast, these observations were not found in DCV and BCV metabolism. To our knowledge, this is the first study to directly demonstrate the effect of CYP3A5*3 genetic variants on the metabolism of ASV. The findings of the present study may provide basic information on ASV, DCV, and BCV metabolisms.
Collapse
Affiliation(s)
- Jun Matsumoto
- Department of Personalized Medicine and Preventive Healthcare Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | - Su Nwe San
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Masachika Fujiyoshi
- Department of Personalized Medicine and Preventive Healthcare Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ayano Kawauchi
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Natsumi Chiba
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Ran Tagai
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Ryoko Sanbe
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Shiho Yanaka
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Hiroaki Sakaue
- Department of Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshinori Kato
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Hiroyoshi Nakamura
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan.,Division of Pharmacy, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Harumi Yamada
- Department of Pharmacokinetics, Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Noritaka Ariyoshi
- Department of Personalized Medicine and Preventive Healthcare Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
17
|
Safar Z, Kis E, Erdo F, Zolnerciks JK, Krajcsi P. ABCG2/BCRP: variants, transporter interaction profile of substrates and inhibitors. Expert Opin Drug Metab Toxicol 2019; 15:313-328. [PMID: 30856014 DOI: 10.1080/17425255.2019.1591373] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION ABCG2 has a broad substrate specificity and is one of the most important efflux proteins modulating pharmacokinetics of drugs, nutrients and toxicokinetics of toxicants. ABCG2 is an important player in transporter-mediated drug-drug interactions (tDDI). Areas covered: The aims of the review are i) to cover transporter interaction profile of substrates and inhibitors that can be utilized to test interaction of drug candidates with ABCG2, ii) to highlight main characteristics of in vitro testing and iii) to describe the structural basis of the broad substrate specificity of the protein. Preclinical data utilizing Abcg2/Bcrp1 knockouts and clinical studies showing effect of ABCG2 c.421C>A polymorphism on pharmacokinetics of drugs have provided evidence for a broad array of drug substrates and support drug - ABCG2 interaction testing. A consensus on using rosuvastatin and sulfasalazine as intestinal substrates for clinical studies is in the formation. Other substrates relevant to the therapeutic area can be considered. Monolayer efflux assays and vesicular transport assays have been extensively utilized in vitro. Expert opinion: Clinical substrates display complex pharmacokinetics due to broad interaction profiles with multiple transporters and metabolic enzymes. Substrate-dependent inhibition has been observed for several inhibitors. Harmonization of in vitro and in vivo testing makes sense. However, rosuvastatin and sulfasalazine are not efficiently transported in either MDCKII or LLC-PK1-based monolayers. Caco-2 monolayer assays and vesicular transport assays are potential alternatives.
Collapse
Affiliation(s)
| | - Emese Kis
- a SOLVO Biotechnology , Szeged , Hungary
| | - Franciska Erdo
- b Faculty of Information Technology and Bionics , Pázmány Péter Catholic University , Budapest , Hungary
| | | | - Peter Krajcsi
- a SOLVO Biotechnology , Szeged , Hungary.,d Department of Morphology and Physiology. Faculty of Health Sciences , Semmelweis University , Budapest , Hungary
| |
Collapse
|
18
|
Giri P, Gupta L, Singh S, Patel N, Srinivas NR, Srivastva BK, Desai RC, Patel PR. Assessment of the in vitro cytochrome P450 (CYP) inhibition potential of ZYTP1, a novel poly (ADP-ribose) polymerase inhibitor. Xenobiotica 2018; 49:1164-1172. [PMID: 30488748 DOI: 10.1080/00498254.2018.1546916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
ZYTP1 is a novel Poly (ADP-ribose) polymerase protein inhibitor being developed for cancer indications. The focus of the work was to determine if ZYTP1 had a perpetrator role in the in vitro inhibition of cytochrome P450 (CYP) enzymes to aid dosing decisions during the clinical development of ZYTP1. ZYTP1 IC50 for CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6 and 3A4/5 was determined using human liver microsomes and LC-MS/MS detection. CYP3A4/5 IC50 of depropylated metabolite of ZYTP1 was also determined. Time dependent inhibition of CYP3A4/5 by ZYTP1 was also assessed using substrates, testosterone and midazolam. The mean IC50 values of ZYTP1 were >100 µM for CYP1A2, 2B6 and 2D6, while 56.1, 24.5, 39.5 and 23.3-58.7 µM for CYP2C8, 2C9, 2C19 and 3A4/5, respectively. The CYP3A4/5 IC50 of depropylated metabolite was 11.95-24.51 µM. Time dependent CYP3A4/5 inhibition was noted for testosterone and midazolam with IC50 shift of 10.9- and 39.9-fold, respectively. With midazolam, the kinact and KI values of ZYTP1 were 0.075 min-1 and 4.47 µM for the CYP3A4/5 time dependent inhibition, respectively. Because of potent inhibition of CYP3A4/5, drugs that undergo metabolism via CYP3A4/5 pathway should be avoided during ZYTP1 therapy.
Collapse
Affiliation(s)
- Poonam Giri
- a Department of Drug Metabolism and Pharmacokinetics , Zydus Research Centre , Ahmadabad , India
| | - Lakshmikant Gupta
- a Department of Drug Metabolism and Pharmacokinetics , Zydus Research Centre , Ahmadabad , India
| | - Sanjay Singh
- a Department of Drug Metabolism and Pharmacokinetics , Zydus Research Centre , Ahmadabad , India
| | - Nirmal Patel
- a Department of Drug Metabolism and Pharmacokinetics , Zydus Research Centre , Ahmadabad , India
| | - Nuggehally R Srinivas
- a Department of Drug Metabolism and Pharmacokinetics , Zydus Research Centre , Ahmadabad , India
| | | | - Ranjit C Desai
- b Department of Medicinal Chemistry , Zydus Research Centre , Ahmadabad , India
| | - Pankaj R Patel
- c Zydus Research Centre , Cadila Healthcare Ltd , Ahmadabad , India
| |
Collapse
|
19
|
Alam K, Crowe A, Wang X, Zhang P, Ding K, Li L, Yue W. Regulation of Organic Anion Transporting Polypeptides (OATP) 1B1- and OATP1B3-Mediated Transport: An Updated Review in the Context of OATP-Mediated Drug-Drug Interactions. Int J Mol Sci 2018. [PMID: 29538325 PMCID: PMC5877716 DOI: 10.3390/ijms19030855] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Organic anion transporting polypeptides (OATP) 1B1 and OATP1B3 are important hepatic transporters that mediate the uptake of many clinically important drugs, including statins from the blood into the liver. Reduced transport function of OATP1B1 and OATP1B3 can lead to clinically relevant drug-drug interactions (DDIs). Considering the importance of OATP1B1 and OATP1B3 in hepatic drug disposition, substantial efforts have been given on evaluating OATP1B1/1B3-mediated DDIs in order to avoid unwanted adverse effects of drugs that are OATP substrates due to their altered pharmacokinetics. Growing evidences suggest that the transport function of OATP1B1 and OATP1B3 can be regulated at various levels such as genetic variation, transcriptional and post-translational regulation. The present review summarizes the up to date information on the regulation of OATP1B1 and OATP1B3 transport function at different levels with a focus on potential impact on OATP-mediated DDIs.
Collapse
Affiliation(s)
- Khondoker Alam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Alexandra Crowe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| | - Xueying Wang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Pengyue Zhang
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kai Ding
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA.
| | - Lang Li
- Center for Computational Biology and Bioinformatics, Indiana Institute of Personalized Medicine, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA.
| | - Wei Yue
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA.
| |
Collapse
|