1
|
Valentin JP, Hoffmann P, Ortemann-Renon C, Koerner J, Pierson J, Gintant G, Willard J, Garnett C, Skinner M, Vargas HM, Wisialowski T, Pugsley MK. The Challenges of Predicting Drug-Induced QTc Prolongation in Humans. Toxicol Sci 2022; 187:3-24. [PMID: 35148401 PMCID: PMC9041548 DOI: 10.1093/toxsci/kfac013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The content of this article derives from a Health and Environmental Sciences Institute (HESI) consortium with a focus to improve cardiac safety during drug development. A detailed literature review was conducted to evaluate the concordance between nonclinical repolarization assays and the clinical thorough QT (TQT) study. Food and Drug Administration and HESI developed a joint database of nonclinical and clinical data, and a retrospective analysis of 150 anonymized drug candidates was reviewed to compare the performance of 3 standard nonclinical assays with clinical TQT study findings as well as investigate mechanism(s) potentially responsible for apparent discrepancies identified. The nonclinical assays were functional (IKr) current block (Human ether-a-go-go related gene), action potential duration, and corrected QT interval in animals (in vivo corrected QT). Although these nonclinical assays demonstrated good specificity for predicting negative clinical QT prolongation, they had relatively poor sensitivity for predicting positive clinical QT prolongation. After review, 28 discordant TQT-positive drugs were identified. This article provides an overview of direct and indirect mechanisms responsible for QT prolongation and theoretical reasons for lack of concordance between clinical TQT studies and nonclinical assays. We examine 6 specific and discordant TQT-positive drugs as case examples. These were derived from the unique HESI/Food and Drug Administration database. We would like to emphasize some reasons for discordant data including, insufficient or inadequate nonclinical data, effects of the drug on other cardiac ion channels, and indirect and/or nonelectrophysiological effects of drugs, including altered heart rate. We also outline best practices that were developed based upon our evaluation.
Collapse
Affiliation(s)
- Jean-Pierre Valentin
- Department of Investigative Toxicology, UCB Biopharma SRL, Braine-l’Alleud B-1420, Belgium
| | | | | | - John Koerner
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | - Jennifer Pierson
- Health and Environmental Sciences Institute, Washington, District of Columbia 20005, USA
| | | | - James Willard
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | - Christine Garnett
- Center for Drug Evaluation and Research, FDA, Silver Spring, Maryland 20993, USA
| | | | - Hugo M Vargas
- Department of Safety Pharmacology & Animal Research Center, Amgen, Thousand Oaks, California 91320, USA
| | - Todd Wisialowski
- Department of Safety Pharmacology, Pfizer, Groton, Connecticut 06340, USA
| | - Michael K Pugsley
- Department of Toxicology, Cytokinetics, South San Francisco, California 94080, USA
| |
Collapse
|
2
|
Ogonowski N, Rukavina Mikusic NL, Kouyoumdzian NM, Choi MR, Fellet A, Balaszczuk AM, Celuch SM. Cardiotoxic Effects of the Antineoplastic Doxorubicin in a Model of Metabolic Syndrome: Oxidative Stress and Transporter Expression in the Heart. J Cardiovasc Pharmacol 2021; 78:784-791. [PMID: 34524257 DOI: 10.1097/fjc.0000000000001137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/18/2021] [Indexed: 12/22/2022]
Abstract
ABSTRACT The aim of the present work was to examine whether metabolic syndrome-like conditions in rats with fructose (F) overload modify the cardiotoxic effects induced by doxorubicin (DOX) and whether the treatment altered the expression of P-gp, breast cancer resistance protein, and organic cation/carnitine transporters in the heart. Male Sprague-Dawley rats received either tap water (control group [C]; n = 16) or water with F 10% wt/vol (n = 16) during 8 weeks. Three days before being killed, the animals received a single dose of DOX (6 mg/kg, ip, md) (C-DOX and F-DOX groups) or vehicle (VEH; ISS 1 mL/kg BW; ip) (C-VEH and F-VEH groups) (n = 8 per group). F overload enhanced thiobarbituric acid-reactive substance levels in the left ventricle, and DOX injection further increased those values. DOX did not alter thiobarbituric acid-reactive substance production in C animals. DOX caused a decrease of 30% in the ejection fraction and a nearly 40% reduction in the fractional shortening in F animals, but not in C rats. Cardiac tissue levels of P-gp decreased by about 30% in F rats compared with the C groups. DOX did not modify cardiac P-gp expression. Breast cancer resistance protein and organic cation/carnitine transporter (OCTN 1/2/3) protein levels did not change with either F or DOX. It is suggested that DOX could cause greater cardiotoxicity in rats receiving F, probably due to enhanced cardiac lipid peroxidation and lower expression of cardiac P-gp. These results support the hypothesis that the cardiotoxicity of DOX could be increased under metabolic syndrome-like conditions or in other health disorders that involve cardiovascular risk factors.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Animals
- Antibiotics, Antineoplastic
- Cardiotoxicity
- Disease Models, Animal
- Doxorubicin
- Heart Diseases/chemically induced
- Heart Diseases/metabolism
- Heart Diseases/pathology
- Heart Diseases/physiopathology
- Lipid Peroxidation
- Male
- Metabolic Syndrome/complications
- Metabolic Syndrome/metabolism
- Myocardium/metabolism
- Myocardium/pathology
- Organic Cation Transport Proteins/genetics
- Organic Cation Transport Proteins/metabolism
- Oxidative Stress
- Rats, Sprague-Dawley
- Ventricular Function, Left/drug effects
- Rats
Collapse
Affiliation(s)
- Natalia Ogonowski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Fisiología, Instituto de Química y Metabolismo del Fármaco, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Lucía Rukavina Mikusic
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Ciudad Autónoma de Buenos Aires, Argentina
| | - Nicolás Martín Kouyoumdzian
- Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET, Ciudad Autónoma de Buenos Aires, Argentina; and
| | - Marcelo Roberto Choi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET, Ciudad Autónoma de Buenos Aires, Argentina; and
| | - Andrea Fellet
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Fisiología, Instituto de Química y Metabolismo del Fármaco, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana María Balaszczuk
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Fisiología, Instituto de Química y Metabolismo del Fármaco, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Stella Maris Celuch
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
3
|
Lee J, Mehrotra S, Zare-Eelanjegh E, Rodrigues RO, Akbarinejad A, Ge D, Amato L, Kiaee K, Fang Y, Rosenkranz A, Keung W, Mandal BB, Li RA, Zhang T, Lee H, Dokmeci MR, Zhang YS, Khademhosseini A, Shin SR. A Heart-Breast Cancer-on-a-Chip Platform for Disease Modeling and Monitoring of Cardiotoxicity Induced by Cancer Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2004258. [PMID: 33094918 PMCID: PMC8049959 DOI: 10.1002/smll.202004258] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/12/2020] [Indexed: 05/02/2023]
Abstract
Cardiotoxicity is one of the most serious side effects of cancer chemotherapy. Current approaches to monitoring of chemotherapy-induced cardiotoxicity (CIC) as well as model systems that develop in vivo or in vitro CIC platforms fail to notice early signs of CIC. Moreover, breast cancer (BC) patients with preexisting cardiac dysfunctions may lead to different incident levels of CIC. Here, a model is presented for investigating CIC where not only induced pluripotent stem cell (iPSC)-derived cardiac tissues are interacted with BC tissues on a dual-organ platform, but electrochemical immuno-aptasensors can also monitor cell-secreted multiple biomarkers. Fibrotic stages of iPSC-derived cardiac tissues are promoted with a supplement of transforming growth factor-β 1 to assess the differential functionality in healthy and fibrotic cardiac tissues after treatment with doxorubicin (DOX). The production trend of biomarkers evaluated by using the immuno-aptasensors well-matches the outcomes from conventional enzyme-linked immunosorbent assay, demonstrating the accuracy of the authors' sensing platform with much higher sensitivity and lower detection limits for early monitoring of CIC and BC progression. Furthermore, the versatility of this platform is demonstrated by applying a nanoparticle-based DOX-delivery system. The proposed platform would potentially help allow early detection and prediction of CIC in individual patients in the future.
Collapse
Affiliation(s)
- Junmin Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Shreya Mehrotra
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Elaheh Zare-Eelanjegh
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Raquel O Rodrigues
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Center for MicroElectromechanical Systems (CMEMS-UMinho), University of Minho, Campus de Azurém, Guimarães, 4800-058, Portugal
| | - Alireza Akbarinejad
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemistry, Faculty of Basic Sciences, Tarbiat Modares University, Tehran, 14115-175, Iran
| | - David Ge
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Luca Amato
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - YongCong Fang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Aliza Rosenkranz
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Wendy Keung
- Dr. Li Dak Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Ronald A Li
- Dr. Li Dak Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong
| | - Ting Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - HeaYeon Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- MARA Nanotech New York, inc., New York, NY, 10031-9101, USA
| | - Mehmet Remzi Dokmeci
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ali Khademhosseini
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
4
|
Effects of Verapamil and Diltiazem on the Pharmacokinetics and Pharmacodynamics of Rivaroxaban. Pharmaceutics 2019; 11:pharmaceutics11030133. [PMID: 30893910 PMCID: PMC6470838 DOI: 10.3390/pharmaceutics11030133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/08/2019] [Accepted: 03/15/2019] [Indexed: 12/29/2022] Open
Abstract
Concomitant use of rivaroxaban with non-dihydropyridine calcium channel blockers (non-DHPs) might lead to an increase of systemic rivaroxaban exposure and anticoagulant effects in relation to the inhibition of metabolic enzymes and/or transporters by non-DHPs. This study was designed to evaluate the effects of verapamil and diltiazem on the pharmacokinetics and the prolongation of prothrombin time of rivaroxaban in rats. The data were analyzed using a pharmacokinetic/pharmacodynamics (PK/PD) modeling approach to quantify the influence of verapamil. Verapamil increased the systemic exposure of rivaroxaban by 2.8-fold (p <0.001) which was probably due to the inhibition of efflux transportation rather than metabolism. Prothrombin time was also prolonged in a proportional manner; diltiazem did not show any significant effects, however. A transit PK model in the absorption process comprehensively describes the double-peaks of rivaroxaban plasma concentrations and the corresponding change of prothrombin time with a simple linear relationship. The slope of prothrombin time vs. rivaroxaban plasma concentration in rats was retrospectively found to be insensitive by about 5.4-fold compared to than in humans. More than a 67% dose reduction in rivaroxaban is suggested in terms of both a pharmacokinetic point of view, and the sensitivity differences on the prolongation of prothrombin time when used concomitantly with verapamil.
Collapse
|
5
|
Sarkaria JN, Hu LS, Parney IF, Pafundi DH, Brinkmann DH, Laack NN, Giannini C, Burns TC, Kizilbash SH, Laramy JK, Swanson KR, Kaufmann TJ, Brown PD, Agar NYR, Galanis E, Buckner JC, Elmquist WF. Is the blood-brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro Oncol 2019; 20:184-191. [PMID: 29016900 DOI: 10.1093/neuonc/nox175] [Citation(s) in RCA: 424] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The blood-brain barrier (BBB) excludes the vast majority of cancer therapeutics from normal brain. However, the importance of the BBB in limiting drug delivery and efficacy is controversial in high-grade brain tumors, such as glioblastoma (GBM). The accumulation of normally brain impenetrant radiographic contrast material in essentially all GBM has popularized a belief that the BBB is uniformly disrupted in all GBM patients so that consideration of drug distribution across the BBB is not relevant in designing therapies for GBM. However, contrary to this view, overwhelming clinical evidence demonstrates that there is also a clinically significant tumor burden with an intact BBB in all GBM, and there is little doubt that drugs with poor BBB permeability do not provide therapeutically effective drug exposures to this fraction of tumor cells. This review provides an overview of the clinical literature to support a central hypothesis: that all GBM patients have tumor regions with an intact BBB, and cure for GBM will only be possible if these regions of tumor are adequately treated.
Collapse
Affiliation(s)
- Jann N Sarkaria
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Leland S Hu
- Mayo Clinic, Scottsdale, Arizona (L.S.H., K.R.S.)
| | - Ian F Parney
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Deanna H Pafundi
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Debra H Brinkmann
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Nadia N Laack
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Caterina Giannini
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Terence C Burns
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Sani H Kizilbash
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Janice K Laramy
- University of Minnesota, Minneapolis, Minnesota (J.K.L., W.F.E.)
| | | | - Timothy J Kaufmann
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Paul D Brown
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | | | - Evanthia Galanis
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - Jan C Buckner
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| | - William F Elmquist
- Mayo Clinic, Rochester, Minnesota (J.N.S., I.F.P., D.H.P., D.H.B., N.N.L., C.G., T.C.B., S.H.K., T.J.K., P.D.B., E.G., J.C.B.)
| |
Collapse
|
6
|
Kim E, Ihm C, Kang W. Modeling of aceclofenac metabolism to major metabolites in healthy volunteers. Drug Metab Pharmacokinet 2016; 31:458-463. [DOI: 10.1016/j.dmpk.2016.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/13/2016] [Accepted: 10/02/2016] [Indexed: 10/20/2022]
|
7
|
Quignot N. Modeling bioavailability to organs protected by biological barriers. In Silico Pharmacol 2013; 1:8. [PMID: 25505653 PMCID: PMC4230447 DOI: 10.1186/2193-9616-1-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 05/05/2013] [Indexed: 01/16/2023] Open
Abstract
Computational pharmacokinetic (PK) modeling gives access to drug concentration vs. time profiles in target organs and allows better interpretation of clinical observations of therapeutic or toxic effects. Physiologically-based PK (PBPK) models in particular, based on mechanistic descriptions of the body anatomy and physiology, may also help to extrapolate in vitro or animal data to human. Once in the systemic circulation, a chemical has access to the microvasculature of every organ or tissue. However, its penetration in the brain, retina, thymus, spinal cord, testis, placenta,… may be limited or even fully prevented by dynamic physiological blood-tissue barriers. Those barriers are both physical (involving tight junctions between adjacent cells) and biochemical (involving metabolizing enzymes and transporters). On those cases, correct mechanistic characterization of the passage (or not) of molecules through the barrier can be crucial for improved PBPK modeling and prediction. In parallel, attempts to understand and quantitatively characterize the processes involved in drug penetration of physiological barriers have led to the development of several in vitro experimental models. Data from such assays are very useful to calibrate PBPK models. We review here those in vitro and computational models, highlighting the challenges and perspectives for in vitro and computational models to better assess drug availability to target tissues.
Collapse
Affiliation(s)
- Nadia Quignot
- Bioengineering Department, Chair of Mathematical Modeling for Systems Toxicology, Université de Technologie de Compiègne, Royallieu Research Center, Compiègne, 60200 France ; LA-SER, Strategy and Decision Analytics, 10 place de la Catalogne, Paris, 75014 France
| |
Collapse
|
8
|
Interactions between bufadienolides derived from toad venom and verapamil in langendorff-perfused guinea-pig hearts. Toxicol In Vitro 2013; 27:396-401. [DOI: 10.1016/j.tiv.2012.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/21/2012] [Accepted: 08/05/2012] [Indexed: 11/17/2022]
|
9
|
Kang W, Elitzer S, Noh K, Bednarek T, Weiss M. Myocardial pharmacokinetics of ebastine, a substrate for cytochrome P450 2J, in rat isolated heart. Br J Pharmacol 2012; 163:1733-9. [PMID: 21410688 DOI: 10.1111/j.1476-5381.2011.01338.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE It is well established that cytochrome P450 2J (CYP2J) enzymes are expressed preferentially in the heart, and that ebastine is a substrate for CYP2J, but it is not known whether ebastine is metabolized in myocardium. Therefore, we investigated its pharmacokinetics in the rat isolated perfused heart. EXPERIMENTAL APPROACH Rat isolated hearts were perfused in the recirculating mode with ebastine for 130 min. The concentrations of ebastine and its metabolites, hydroxyebastine and carebastine, were measured using liquid chromatography with a tandem mass spectrometry. The data were analysed by a compartmental model. The time course of negative inotropic response was linked to ebastine concentration to determine the concentration-effect relationship. KEY RESULTS Ebastine was metabolized to an intermediate compound, hydroxyebastine, which was subsequently further metabolized to carebastine. No desalkylebastine was found. The kinetics of the sequential metabolism of ebastine was well described by the compartmental model. The EC(50) of the negative inotropic effect of ebastine in rat isolated heart was much higher than free plasma concentrations in humans after clinical doses. CONCLUSIONS AND IMPLICATIONS The kinetics of ebastine conversion to carebastine via hydroxyebastine resembled that observed in human liver microsomes. The results may be of interest for functional characterization of CYP2J activity in rat heart.
Collapse
Affiliation(s)
- W Kang
- College of Pharmacy, Yeungnam University, Kyoungbuk, South Korea Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | | | | | | |
Collapse
|
10
|
Weiss M. Functional characterization of drug uptake and metabolism in the heart. Expert Opin Drug Metab Toxicol 2011; 7:1295-306. [DOI: 10.1517/17425255.2011.614233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
11
|
Kim Y, Park K, Kang W. Effect of fluvastatin, lovastatin, nifedipine and verapamil on the systemic exposure of nateglinide in rabbits. Biopharm Drug Dispos 2010; 31:443-9. [DOI: 10.1002/bdd.724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Pharmacokinetic evaluation and modeling of formulated levodopa intranasal delivery systems. Eur J Pharm Sci 2009; 38:525-32. [DOI: 10.1016/j.ejps.2009.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 09/07/2009] [Accepted: 09/27/2009] [Indexed: 11/17/2022]
|
13
|
Sermsappasuk P, Weiss M. Modelling of alpha1-adrenoceptor-mediated temporal dynamics of inotropic response in rat heart to assess ligand binding and signal transduction parameters. Br J Pharmacol 2009; 156:764-73. [PMID: 19220295 DOI: 10.1111/j.1476-5381.2008.00013.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE In order to use the transient response to an antagonist (prazosin) to evaluate properties of agonist interactions with the alpha(1)-adrenoceptor system, an integrative mechanistic model of cardiac uptake of prazosin and its competitive interaction with phenylephrine at the receptor site was developed. Based on the operational model of agonism, the aim was to evaluate both the receptor binding and signal transduction process as determinants of the inotropic effect of phenylephrine. EXPERIMENTAL APPROACH In Langendorff-perfused rat hearts, prazosin outflow concentration and left ventricular developed pressure were measured, first in the presence of 12.3 micromol x L(-1) phenylephrine following a 1 min infusion of 1.27 nmol [(3)H]-prazosin, and second, when after 30 min the phenylephrine concentration in perfusate was reduced to 6.1 micromol x L(-1), the 1 min infusion of 1.27 nmol [(3)H]-prazosin was repeated. KEY RESULTS The kinetic model accounted for cardiac uptake and receptor binding kinetics of prazosin (dissociation constant, mean +/- SD: 0.057 +/- 0.012 nmol.L(-1)), assuming that the competitive displacement of phenylephrine (dissociation constant: 101 +/- 13 nmol x L(-1)) reduced the receptor occupation by the agonist and, consequently, contractility. This competitive binding process appeared to be the rate-determining step in response generation. The relationship between receptor occupancy and inotropic response was described by an efficacy parameter (tau, ratio of receptor density and coupling efficiency) of 4.9. CONCLUSIONS AND IMPLICATIONS Mechanistic pharmacodynamic modelling of the kinetics of antagonism by prazosin allows quantitative assessment of the alpha(1)-adrenoceptor system both at the receptor and post-receptor levels.
Collapse
Affiliation(s)
- P Sermsappasuk
- Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | |
Collapse
|
14
|
Audi SH, Merker MP, Krenz GS, Ahuja T, Roerig DL, Bongard RD. Coenzyme Q1 redox metabolism during passage through the rat pulmonary circulation and the effect of hyperoxia. J Appl Physiol (1985) 2008; 105:1114-26. [PMID: 18703762 DOI: 10.1152/japplphysiol.00177.2008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The objective was to evaluate the pulmonary disposition of the ubiquinone homolog coenzyme Q(1) (CoQ(1)) on passage through lungs of normoxic (exposed to room air) and hyperoxic (exposed to 85% O(2) for 48 h) rats. CoQ(1) or its hydroquinone (CoQ(1)H(2)) was infused into the arterial inflow of isolated, perfused lungs, and the venous efflux rates of CoQ(1)H(2) and CoQ(1) were measured. CoQ(1)H(2) appeared in the venous effluent when CoQ(1) was infused, and CoQ(1) appeared when CoQ(1)H(2) was infused. In normoxic lungs, CoQ(1)H(2) efflux rates when CoQ(1) was infused decreased by 58 and 33% in the presence of rotenone (mitochondrial complex I inhibitor) and dicumarol [NAD(P)H-quinone oxidoreductase 1 (NQO1) inhibitor], respectively. Inhibitor studies also revealed that lung CoQ(1)H(2) oxidation was via mitochondrial complex III. In hyperoxic lungs, CoQ(1)H(2) efflux rates when CoQ(1) was infused decreased by 23% compared with normoxic lungs. Based on inhibitor effects and a kinetic model, the effect of hyperoxia could be attributed predominantly to 47% decrease in the capacity of complex I-mediated CoQ(1) reduction, with no change in the other redox processes. Complex I activity in lung homogenates was also lower for hyperoxic than for normoxic lungs. These studies reveal that lung complexes I and III and NQO1 play a dominant role in determining the vascular concentration and redox status of CoQ(1) during passage through the pulmonary circulation, and that exposure to hyperoxia decreases the overall capacity of the lung to reduce CoQ(1) to CoQ(1)H(2) due to a depression in complex I activity.
Collapse
Affiliation(s)
- Said H Audi
- Research Service 151, Zablocki VAMC, 5000 W. National Avenue, Milwaukee, WI 53295, USA.
| | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Abstract
A major part of the science of pharmacokinetics is the modeling of the underlying processes that contribute to drug disposition. The purpose of pharmacokinetic models is to summarize the knowledge gained in preclinical and clinical studies at various stages in drug development and to rationally guide future studies with the use of adequately predictive models. This review highlights a variety of recent advances in mechanistic pharmacokinetic modeling. It is aimed at a broad audience, and hence, an attempt was made to maintain a balance between technical information and practical applications of pharmacokinetic modeling. It is hoped that drug researchers from all disciplines would be able to get a flavor of the function and capacity of pharmacokinetic modelers and their contribution to drug development. While this review is not intended to be a technical reference on modeling approaches, the roles of statistical applications and population methodologies are discussed where appropriate.
Collapse
Affiliation(s)
- Alaa M Ahmad
- Department of Clinical Pharmacology, Vertex Pharmaceuticals Inc., 130 Waverly Street, Cambridge, MA 02139, USA.
| |
Collapse
|
17
|
Yun HY, Park HC, Kang W, Kwon KI. Pharmacokinetic and pharmacodynamic modelling of the effects of glimepiride on insulin secretion and glucose lowering in healthy humans. J Clin Pharm Ther 2006; 31:469-76. [PMID: 16958825 DOI: 10.1111/j.1365-2710.2006.00766.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glimepiride is an oral sulfonylurea antihyperglycaemic agent. We used pharmacokinetic-pharmacodynamic (PK-PD) modelling to analyse the relationship between plasma glimepiride concentration, insulin secretion and glucose lowering to determine the effects of the drug in healthy volunteers. A single 2-mg oral dose of glimepiride was administered to six healthy volunteers. The control group received a placebo. All subjects consumed 12 g of sugar immediately after drug administration in order to standardize the initial plasma glucose levels. Serial blood sampling was performed for 9 h after oral dosing. Plasma glimepiride, insulin and glucose levels were determined by validated methods (LC/MS/MS assay, hexokinase method and radioimmunoassay respectively). Time courses of plasma glimepiride concentration, insulin secretion, and glucose lowering effects were analysed by means of PK-PD modelling with the ADAPT II program. The time course of the plasma concentrations followed a two-compartmental model with a lag time. The glimepiride concentration peaked at 191.5 ng/mL at approximately 4 h after administration. The maximal increase in insulin secretion was 9.98 mIU/L and the maximal decrease in plasma glucose was 19.33 mg/dL. Both peak effects occurred at approximately 2.5 h after drug intake. The glucose disappearance model was used to analyse glimepiride's insulin secretion and glucose lowering effects. The PK-PD model described well the relationship between plasma glimepiride and its insulin secretion and hypoglycaemic effects in healthy volunteers.
Collapse
Affiliation(s)
- H-Y Yun
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | | | | | | |
Collapse
|
18
|
Sermsappasuk P, Abdelrahman O, Weiss M. Modeling Cardiac Uptake and Negative Inotropic Response of Verapamil in Rat Heart: Effect of Amiodarone. Pharm Res 2006; 24:48-57. [PMID: 16969694 DOI: 10.1007/s11095-006-9117-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 07/10/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE To determine the effect of the P-glycoprotein (Pgp) modulator amiodarone on the pharmacokinetics and pharmacodynamics (PK/PD) of Pgp substrate verapamil in the perfused rat heart. METHODS In Langendorff-perfused rat hearts, the outflow concentration-time curve and inotropic response data were measured after a 1.5 nmol dose of [3H]-verapamil (infused within 1 min) in the absence and presence of the amiodarone (1 microM) in perfusate, as well as using a double dosing regimen (0.75 nmol in a 10 min interval). These data were analyzed by a PK/PD model. RESULTS Amiodarone failed to influence the rapid uptake and equilibrium partitioning of verapamil into the heart. The time course of the negative inotropic effect of verapamil, including the 'rebound' above the original baseline after the infusion of verapamil was stopped, could be described by a PK/PD tolerance model. Tolerance development (mean delay time, 12 min) led to a reduction in predicted steady-state effect (16%). The EC50 and Emax values as estimated in single dose experiments were 16.4+/-4.1 nM and 50.5+/-18.9 mmHg, respectively. CONCLUSIONS The result does not support the hypothesis that Pgp inhibition by amiodarone increases cardiac uptake of the Pgp substrate verapamil.
Collapse
Affiliation(s)
- Pakawadee Sermsappasuk
- Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenberg, D-06097, Halle, Germany
| | | | | |
Collapse
|
19
|
Dakhel Y, Jamali F. Erythromycin potentiates PR interval prolonging effect of verapamil in the rat: A pharmacodynamic drug interaction. Toxicol Appl Pharmacol 2006; 214:24-9. [PMID: 16466760 DOI: 10.1016/j.taap.2005.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Revised: 11/03/2005] [Accepted: 11/26/2005] [Indexed: 11/25/2022]
Abstract
Calcium channel blockers and macrolide antibiotics account for many drug interactions. Anecdotal reports suggest interactions between the two resulting in severe side effects. We studied the interaction between verapamil and erythromycin in the rat to see whether it occurs at the pharmacokinetics or pharmacodynamic level. Adult male Sprague-Dawley rats received doses of 1 mg/kg verapamil or 100 mg/kg erythromycin alone or in combination (n = 6/group). Serial blood samples (0-6 h) were taken for determination of the drug concentrations using HPLC. Electrocardiograms were recorded (0-6 h) through subcutaneously inserted lead II. Binding of the drugs to plasma proteins was studied using spiked plasma. Verapamil prolonged PR but not QT interval. Erythromycin prolonged QT but not PR interval. The combination resulted in a significant increase in PR interval prolongation and AV node blocks but did not further prolong QT interval. Pharmacokinetics and protein binding of neither drug were altered by the other. Our rat data confirm the anecdotal human case reports that combination of erythromycin and verapamil can result in potentiation of the cardiovascular response. The interaction appears to be at the pharmacodynamic rather than pharmacokinetic level hence may be extrapolated to other calcium channel antagonists.
Collapse
Affiliation(s)
- Yaman Dakhel
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada T6H 2N8
| | | |
Collapse
|
20
|
Weiss M, Giessler C, Kang W. Effects of idarubicin and idarubicinol on rat coronary resistance and vasoconstrictor responsiveness of isolated aorta and mesentery. Anticancer Drugs 2006; 17:69-74. [PMID: 16317292 DOI: 10.1097/01.cad.0000185186.03099.31] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
It has been hypothesized that coronary vasoconstriction is involved in the cardiotoxic action of anthracyclines. The purpose of this study was to determine whether an increase in coronary resistance induced by idarubicin (IDA) or its primary circulating metabolite idarubicinol (IDOL) is correlated with a decrease in vascular sensitivity to vasoconstrictor agonists. Coronary vasoconstriction was studied in single-pass perfused rat hearts after a 10-min infusion of 0.5 mg IDA or IDOL. In the endothelium-intact rat thoracic aorta and mesentery we measured the inhibition of phenylephrine (PE)- and KCl-induced contraction in the presence of IDA and IDOL, respectively. The increase in coronary vascular resistance evoked by IDOL (121%) exceeded that of IDA (75%). IDA (10-100 micromol/l) concentration-dependently diminished vascular sensitivity to PE and KCl due to a reduction in maximal contractile response (Emax), i.e. the antagonism by IDA of PE- or KCl-induced vasoconstriction was non-competitive, indicating a post-receptor cellular mechanism. These reductions of PE or KCl efficacy elicited by IDOL were significantly larger than those elicited by the corresponding doses of IDA. The decrease in efficacy of PE in the presence of IDA and IDOL was characterized by IC50 estimates of 44.3 and 30.7 mumol/l, respectively. With a 10-fold lower IC50, IDA inhibited the reactivity of small mesenteric arteries to noradrenaline with 10-fold higher potency. The correlation between the increase in coronary resistance and the decrease in vasoconstrictor responsiveness may suggest that these anthracyclines act through a common cellular mechanism.
Collapse
Affiliation(s)
- Michael Weiss
- Section of Pharmacokinetics, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | | | | |
Collapse
|
21
|
Hoffmann P, Warner B. Are hERG channel inhibition and QT interval prolongation all there is in drug-induced torsadogenesis? A review of emerging trends. J Pharmacol Toxicol Methods 2005; 53:87-105. [PMID: 16289936 DOI: 10.1016/j.vascn.2005.07.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 07/26/2005] [Indexed: 11/21/2022]
Abstract
Contemporary preclinical in vitro and in vivo methods have been imperfect in predicting drug-induced Torsades de Pointes (TdP) in humans. A better understanding of additional relevant factors in the genesis of drug-induced TdP is necessary. New sophisticated in vitro techniques, such as arterially perfused ventricular wedge preparations or isolated perfused hearts, potentially offer a better understanding of torsadogenic mechanisms and a refinement of drug testing. Of particular interest are the dispersion of repolarization and the refractoriness of different cell types across the ventricular wall, triangulation of the action potential, reverse use dependence and instability of the action potential duration. In vivo models are currently refined by establishing parameters such as beat-to-beat variability and T-wave morphology as derived from the in vitro proarrhythmia indices. Animal models of proarrhythmia are to date not recommended for routine evaluation. A pharmacodynamic interaction with combinations of torsadogenic compounds is another area to be considered. Little is known about channel/receptor cross talk, although considerable evidence exists that cardiac G protein-coupled receptors can modulate hERG channel function. More investigations are necessary to further evaluate the role of altered gene expression, mutations, and polymorphisms in drug-induced TdP. A novel mechanism of drug-induced torsadogenesis is the reduced expression of hERG channel protein on the plasma membrane due to a trafficking defect. Pharmacokinetic and metabolism data are crucial for calculating the risk of a torsadogenic potential in man. Consideration of intracardiac accumulation can help in delineating pharmacokinetic-pharmacodyamic relationships. In silico virtual screening procedures with new chemical entities to predict hERG block may develop as a promising tool. The role of in silico modeling of TdP arrhythmia is likely to become increasingly important for organizing and integrating the vast amount of generated data. At present, however, in silico methods cannot replace existing preclinical in vitro and in vivo models.
Collapse
Affiliation(s)
- Peter Hoffmann
- Safety and Technical Sciences, F. Hoffmann-La Roche Ltd, 4070 Basel, Switzerland.
| | | |
Collapse
|
22
|
Balayssac D, Authier N, Cayre A, Coudore F. Does inhibition of P-glycoprotein lead to drug–drug interactions? Toxicol Lett 2005; 156:319-29. [PMID: 15763631 DOI: 10.1016/j.toxlet.2004.12.008] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2004] [Revised: 12/22/2004] [Accepted: 12/22/2004] [Indexed: 01/12/2023]
Abstract
Permeability-glycoprotein (Pgp) is a drug transporter responsible for the efflux of xenobiotics out of cells that influence the pharmacokinetics of numerous drugs. However, the role of this transporter in drug-drug interactions is still poorly studied even though a lot of P-glycoprotein substrates and P-glycoprotein inhibitors are identified among drugs of standard usage. On one hand, Pgp is distributed within a lot of organs and tissues implicated in the absorption or excretion of xenobiotics, and drug-drug interactions with this protein may increase the bioavailability of simultaneously administered active drugs. On the other hand, Pgp is linked to the integrity of blood-tissue barriers, such as the blood-brain barrier or placenta, and a partial blockage of Pgp could be responsible for a new drug distribution in the organism with possible increase of drug rates in organs behind these barriers. Therefore, concomitant administration of substrates and Pgp inhibitors would modify drug pharmacokinetics by increasing bioavailability and organ uptake, leading to more adverse drug reactions and toxicities. Consequently, the identification and comprehension of these drug-drug interactions remain important keys to risk assessment.
Collapse
Affiliation(s)
- David Balayssac
- Laboratory of Toxicology, Faculty of Pharmacy, CHU G. Montpied, 28 Place Henri Dunant, 63000 Clermont-Ferrand, France.
| | | | | | | |
Collapse
|
23
|
Roerig DL, Audi SH, Ahlf SB. KINETIC CHARACTERIZATION OF P-GLYCOPROTEIN-MEDIATED EFFLUX OF RHODAMINE 6G IN THE INTACT RABBIT LUNG. Drug Metab Dispos 2004; 32:953-8. [PMID: 15319336 DOI: 10.1124/dmd.104.000042] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
P-Glycoprotein (P-gp) is an ATP-dependent drug efflux transporter involved in multidrug resistance and drug disposition in many organ systems. A majority of P-gp substrates are lipophilic amine drugs which also exhibit rapid extensive accumulation in lung tissue. P-gp is expressed in lung tissue, and the very nature of this drug efflux mechanism suggests a moderating role in pulmonary drug disposition. Little is known about P-gp-mediated efflux out of lung tissue or its kinetic characteristics as they may relate to the impact of P-gp on pulmonary drug accumulation. The present study develops an experimental and kinetic model to characterize the kinetics of P-gp-mediated efflux of rhodamine 6G dye (R6G) out of the intact rabbit lung. The perfusate concentration of R6G with time during recirculation through an isolated perfused rabbit lung was measured, and 66.6 +/- 2.6% (S.E.) of the perfusate R6G was taken up by the lung. In the presence of P-gp inhibitors, R6G uptake increased significantly to 87.5 +/- 1.1% (P < 0.002), indicating a functional pulmonary P-gp efflux transporter. Fractional lung accumulation of R6G increased with increasing R6G perfusate concentration, a result consistent with saturation of an efflux transporter. A parsimonious three-compartment kinetic model of R6G pulmonary disposition was used to interpret data sets from experiments with different perfusion variables and to estimate parameters descriptive of the dominant kinetic processes involved in R6G pulmonary accumulation. The estimated value of the kinetic parameter, k(pgp), rate constant for P-gp-mediated R6G efflux, indicates that this transporter plays a significant role in moderating R6G pulmonary disposition.
Collapse
Affiliation(s)
- David L Roerig
- Research Service 151, V.A. Medical Center, 5000 W. National Ave., Milwaukee, WI 53295, USA.
| | | | | |
Collapse
|
24
|
|
25
|
Kuhlmann O, Hofmann HS, Müller SP, Weiss M. Pharmacokinetics of idarubicin in the isolated perfused rat lung: effect of cinchonine and rutin. Anticancer Drugs 2003; 14:411-6. [PMID: 12853881 DOI: 10.1097/00001813-200307000-00004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This study was designed to examine the effect of rutin and cinchonine on the uptake and metabolism of idarubicin (IDA) in the isolated perfused rat lung. IDA (2 mg) was infused for 2 min into the truncus pulmonalis in the presence of P-glycoprotein (P-gp) modulators cinchonine (1 microM) or rutin (6 microM). (Rutin is also known as an aldo-keto reductase inhibitor.) Venous outflow samples were collected up to 60 min, and the concentration of IDA and its primary metabolite idarubicinol (IDOL) were measured by high-performance liquid chromatography) with fluorescence detection. Thereafter, the tissue concentrations of IDA and IDOL were determined in the lung (n = 5 in each group). The estimated mean transit times for IDA in the treatment groups (MTT(cinchonine) = 21.8+/-3.5 min; MTT(rutin) = 20.1+/-5.0 min) were significantly higher than in the control group (11.6+/-2.1 min). Both cinchonine and rutin significantly enhanced the lung tissue concentrations of IDA (1.7- and 2.4-fold), as well as of IDOL (2.1- and 2.4-fold). Cinchonine and rutin also increased the outflow recovery of IDOL 2.6- and 2.7-fold, respectively. The results suggest that uptake kinetics of IDA into the rat lung is partly controlled by a P-gp efflux pump and its inhibition enhances the accumulation of IDA.
Collapse
Affiliation(s)
- Olaf Kuhlmann
- Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenberg, Germany
| | | | | | | |
Collapse
|
26
|
Kang W, Weiss M. Modeling the metabolism of idarubicin to idarubicinol in rat heart: effect of rutin and phenobarbital. Drug Metab Dispos 2003; 31:462-8. [PMID: 12642473 DOI: 10.1124/dmd.31.4.462] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Since the severe cardiotoxicity of anthracyclines has been attributed to the intramyocardial formation of C-13 alcohol metabolites, the kinetics of cardiac metabolite formation and disposition as well as the effect of carbonyl reductase inhibitors are of specific interest. This study was designed to investigate the effect of rutin and phenobarbital on the pharmacokinetics of idarubicin (IDA) and its conversion to idarubicinol (IDOL) in the single-pass perfused rat heart. After infusion of IDA (0.5 mg) during 1min, the venous outflow concentrations of IDA and IDOL were measured up to 80 min in the presence and absence of rutin and phenobarbital. A kinetic model was developed to help to interpret the concentration profiles in terms of compartmentation of IDOL formation and to estimate parameters quantitatively descriptive of the transport and biotransformation processes. Rutin and phenobarbital significantly reduced the residual amount of IDOL in heart to 64 and 47% of control, respectively. Pharmacokinetic modeling of the data revealed that IDOL is generated in two different compartments, besides the tissue compartment characterized by saturable uptake, also the compartment that accounts for the quasi-instantaneous initial distribution process is involved. The efflux rate constant of IDOL, k(21,IDOL,) was much smaller than that of IDA. Rutin and phenobarbital significantly reduced IDOL production. Additionally, phenobarbital competitively inhibited the saturable uptake of both IDA and IDOL (increase in apparent Michaelis constants). Reanalysis of data obtained in previous experiments showed that P-glycoprotein inhibitors (verapamil and amiodarone) reduced IDOL uptake in a similar way as already shown for IDA. The present study further supports the utility of pharmacokinetic modeling in identifying sites of drug interactions within the heart.
Collapse
Affiliation(s)
- Wonku Kang
- Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenberg, 06097 Halle, Germany
| | | |
Collapse
|
27
|
Kang W, Weiss M. Digoxin uptake, receptor heterogeneity, and inotropic response in the isolated rat heart: a comprehensive kinetic model. J Pharmacol Exp Ther 2002; 302:577-83. [PMID: 12130718 DOI: 10.1124/jpet.302.2.577] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cardiac pharmacokinetics of digitalis glycosides is not well understood. In the present study, a mechanism-based pharmacokinetic/pharmacodynamic model was developed to describe the uptake kinetics, receptor interaction, and positive inotropic effect of digoxin in the single-pass isolated perfused rat heart. Three doses of digoxin (0.1, 0.2, and 0.3 micromol) were administered to the heart (n = 12) as consecutive 1-min infusions followed by 15-min washout periods. Outflow concentration and left ventricular developed pressure were measured and analyzed by the model. The uptake of digoxin by the heart was limited by capillary permeability with a permeation clearance of 2.35 ml/min/g (about one-third of perfusate flow). Binding kinetics was determined by a mixture of two receptor subtypes, a low-affinity/high-capacity binding site (K(D,1) = 20.9 nmol, 89% of total receptors) and a high-affinity/low-capacity binding site (K(D,2) = 1.5 nmol, 11%). The time course of inotropic response was linked to receptor occupation, with higher efficiency of the high-affinity receptor population. The results suggest that, in the rat heart, consecutive inhibition of first the alpha(2)- and then the alpha(1)-isoform of Na(+)/K(+)-ATPase mediates the positive inotropic effect of digoxin with increasing dosage.
Collapse
Affiliation(s)
- Wonku Kang
- Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenburg, 06097 Halle, Germany
| | | |
Collapse
|