1
|
Lewis JH, Korkmaz SY, Rizk CA, Copeland MJ. Diagnosis, prevention and risk-management of drug-induced liver injury due to medications used to treat mycobacterium tuberculosis. Expert Opin Drug Saf 2024; 23:1093-1107. [PMID: 39212296 DOI: 10.1080/14740338.2024.2399074] [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: 04/16/2024] [Revised: 07/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Many of the first line medications for the treatment of active and latent M. tuberculosis are hepatoxic and cause a spectrum of anti-tuberculosis drug induced liver injury (ATLI), including acute liver failure (ALF). Despite advances in recognition of and prevention of ATLI, isoniazid remains one of the leading causes of DILI as well as drug-induced ALF. AREAS COVERED A literature search of the incidence, risk factors, current societal guidelines, monitoring, and prophylactic medication usage in ATLI was performed using PubMed and institutional websites. Relevant articles from 1972 to 2024 were included in this review. EXPERT OPINION Current societal guidelines regarding ATLI monitoring are mixed, but many recommend liver enzyme testing of high-risk populations. We recommend liver test monitoring for all patients on multi-drug therapy as well as those on isoniazid therapy. Precision medicine practices, such as N-acetyltransferase-2 polymorphism genotyping, are thought to be beneficial in reducing the incidence of ATLI in high-risk populations. However, broader implementation is currently cost prohibitive. Hepatoprotective drugs are not currently recommended, although we do recognize their potential. In patients who develop ATLI but require ongoing anti-TB treatment, strategies to restart the same or less hepatotoxic regimens are currently being followed.
Collapse
Affiliation(s)
- James H Lewis
- Department of Medicine, Division of Gastroenterology-Hepatology, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Serena Y Korkmaz
- Department of Medicine, General Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Courtney A Rizk
- Department of Medicine, General Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Matthew J Copeland
- Department of Medicine, Division of Infectious Diseases, Washington, DC, USA
| |
Collapse
|
2
|
Shan Z, Yang X, Liu H, Yuan Y, Xiao Y, Nan J, Zhang W, Song W, Wang J, Wei F, Zhang Y. Cryo-EM structures of human organic anion transporting polypeptide OATP1B1. Cell Res 2023; 33:940-951. [PMID: 37674011 PMCID: PMC10709409 DOI: 10.1038/s41422-023-00870-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/17/2023] [Indexed: 09/08/2023] Open
Abstract
Members of the solute carrier organic anion transporting polypeptide (OATPs) family function as transporters for a large variety of amphipathic organic anions including endogenous metabolites and clinical drugs, such as bile salts, steroids, thyroid hormones, statins, antibiotics, antivirals, and anticancer drugs. OATP1B1 plays a vital role in transporting such substances into the liver for hepatic clearance. FDA and EMA recommend conducting in vitro testing of drug-drug interactions (DDIs) involving OATP1B1. However, the structure and working mechanism of OATPs still remains elusive. In this study, we determined cryo-EM structures of human OATP1B1 bound with representative endogenous metabolites (bilirubin and estrone-3-sulfate), a clinical drug (simeprevir), and a fluorescent indicator (2',7'-dichlorofluorescein), in both outward- and inward-open states. These structures reveal major and minor substrate binding pockets and conformational changes during transport. In combination with mutagenesis studies and molecular dynamics simulations, our work comprehensively elucidates the transport mechanism of OATP1B1 and provides the structural basis for DDI predictions involving OATP1B1, which will greatly promote our understanding of OATPs.
Collapse
Affiliation(s)
- Ziyang Shan
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xuemei Yang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Huihui Liu
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yafei Yuan
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuan Xiao
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jing Nan
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wenqi Song
- Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jufang Wang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Feiwen Wei
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yanqing Zhang
- Shanghai Fifth People's Hospital, Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Miners JO, Polasek TM, Hulin JA, Rowland A, Meech R. Drug-drug interactions that alter the exposure of glucuronidated drugs: Scope, UDP-glucuronosyltransferase (UGT) enzyme selectivity, mechanisms (inhibition and induction), and clinical significance. Pharmacol Ther 2023:108459. [PMID: 37263383 DOI: 10.1016/j.pharmthera.2023.108459] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Drug-drug interactions (DDIs) arising from the perturbation of drug metabolising enzyme activities represent both a clinical problem and a potential economic loss for the pharmaceutical industry. DDIs involving glucuronidated drugs have historically attracted little attention and there is a perception that interactions are of minor clinical relevance. This review critically examines the scope and aetiology of DDIs that result in altered exposure of glucuronidated drugs. Interaction mechanisms, namely inhibition and induction of UDP-glucuronosyltransferase (UGT) enzymes and the potential interplay with drug transporters, are reviewed in detail, as is the clinical significance of known DDIs. Altered victim drug exposure arising from modulation of UGT enzyme activities is relatively common and, notably, the incidence and importance of UGT induction as a DDI mechanism is greater than generally believed. Numerous DDIs are clinically relevant, resulting in either loss of efficacy or an increased risk of adverse effects, necessitating dose individualisation. Several generalisations relating to the likelihood of DDIs can be drawn from the known substrate and inhibitor selectivities of UGT enzymes, highlighting the importance of comprehensive reaction phenotyping studies at an early stage of drug development. Further, rigorous assessment of the DDI liability of new chemical entities that undergo glucuronidation to a significant extent has been recommended recently by regulatory guidance. Although evidence-based approaches exist for the in vitro characterisation of UGT enzyme inhibition and induction, the availability of drugs considered appropriate for use as 'probe' substrates in clinical DDI studies is limited and this should be research priority.
Collapse
Affiliation(s)
- John O Miners
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Thomas M Polasek
- Certara, Princeton, NJ, USA; Centre for Medicines Use and Safety, Monash University, Melbourne, Australia
| | - Julie-Ann Hulin
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Andrew Rowland
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Robyn Meech
- Discipline of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, Flinders University College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
4
|
Okubo H, Atsukawa M, Okubo T, Ando H, Nakadera E, Ikejima K, Nagahara A. Gadoxetic acid-enhanced magnetic resonance imaging predicts hyperbilirubinemia induced by glecaprevir during hepatitis C virus treatment. Sci Rep 2022; 12:7847. [PMID: 35552472 PMCID: PMC9098462 DOI: 10.1038/s41598-022-11707-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
Glecaprevir is a substrate for organic anion-transporting polypeptide (OATP) 1B1/1B3, which transports bilirubin. Hyperbilirubinemia is an adverse event during anti-hepatitis C virus treatment with glecaprevir and pibrentasvir. Gadoxetic acid is also transported by OATP1B1/1B3, and we aimed to evaluate whether gadoxetic acid-enhanced magnetic resonance (MR) imaging was associated with glecaprevir trough concentrations (Ctrough). We further determined whether this was predictive of hyperbilirubinemia development in a cohort of 33 patients. The contrast enhancement index (CEI), a measure of hepatic enhancement effect on the hepatobiliary image, was assessed. Glecaprevir Ctrough was determined 7 days after administration. Five of the 33 patients (15%) developed Common Terminology Criteria for Adverse Events grade ≥ 2 hyperbilirubinemia. We found a negative relationship between CEI and Ctrough (r = − 0.726, p < 0.001). The partial correlation coefficient between CEI and Ctrough was − 0.654 (p < 0.001), while excluding the effects of albumin, FIB-4 index, and indirect bilirubin at baseline. The Ctrough was significantly higher in patients with hyperbilirubinemia than in those without (p = 0.008). In multivariate analysis, CEI ≤ 1.71 was an independent factor influencing the development of hyperbilirubinemia (p = 0.046). Our findings indicate that gadoxetic acid MR imaging can help predict glecaprevir concentration and development of hyperbilirubinemia.
Collapse
Affiliation(s)
- Hironao Okubo
- Department of Gastroenterology, Juntendo University Nerima Hospital, 3-1-10 Takanodai, Nerima-ku, Tokyo, 177-8521, Japan.
| | - Masanori Atsukawa
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamagari, Inzai, Chiba, 270-1694, Japan.,Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan
| | - Tomomi Okubo
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School Chiba Hokusoh Hospital, 1715 Kamagari, Inzai, Chiba, 270-1694, Japan
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Eisuke Nakadera
- Department of Gastroenterology, Juntendo University Nerima Hospital, 3-1-10 Takanodai, Nerima-ku, Tokyo, 177-8521, Japan
| | - Kenichi Ikejima
- Department of Gastroenterology, Graduate School of Medicine, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Akihito Nagahara
- Department of Gastroenterology, Graduate School of Medicine, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
5
|
Li Y, Talebi Z, Chen X, Sparreboom A, Hu S. Endogenous Biomarkers for SLC Transporter-Mediated Drug-Drug Interaction Evaluation. Molecules 2021; 26:5500. [PMID: 34576971 PMCID: PMC8466752 DOI: 10.3390/molecules26185500] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 12/31/2022] Open
Abstract
Membrane transporters play an important role in the absorption, distribution, metabolism, and excretion of xenobiotic substrates, as well as endogenous compounds. The evaluation of transporter-mediated drug-drug interactions (DDIs) is an important consideration during the drug development process and can guide the safe use of polypharmacy regimens in clinical practice. In recent years, several endogenous substrates of drug transporters have been identified as potential biomarkers for predicting changes in drug transport function and the potential for DDIs associated with drug candidates in early phases of drug development. These biomarker-driven investigations have been applied in both preclinical and clinical studies and proposed as a predictive strategy that can be supplanted in order to conduct prospective DDIs trials. Here we provide an overview of this rapidly emerging field, with particular emphasis on endogenous biomarkers recently proposed for clinically relevant uptake transporters.
Collapse
Affiliation(s)
| | | | | | | | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (Y.L.); (Z.T.); (X.C.); (A.S.)
| |
Collapse
|
6
|
Te Brake LHM, de Jager V, Narunsky K, Vanker N, Svensson EM, Phillips PPJ, Gillespie SH, Heinrich N, Hoelscher M, Dawson R, Diacon AH, Aarnoutse RE, Boeree MJ. Increased bactericidal activity but dose-limiting intolerability at 50 mg·kg -1 rifampicin. Eur Respir J 2021; 58:13993003.00955-2020. [PMID: 33542056 PMCID: PMC8411896 DOI: 10.1183/13993003.00955-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 12/07/2020] [Indexed: 01/16/2023]
Abstract
Background Accumulating data indicate that higher rifampicin doses are more effective and shorten tuberculosis (TB) treatment duration. This study evaluated the safety, tolerability, pharmacokinetics, and 7- and 14-day early bactericidal activity (EBA) of increasing doses of rifampicin. Here we report the results of the final cohorts of PanACEA HIGHRIF1, a dose escalation study in treatment-naive adult smear-positive patients with TB. Methods Patients received, in consecutive cohorts, 40 or 50 mg·kg−1 rifampicin once daily in monotherapy (day 1–7), supplemented with standard dose isoniazid, pyrazinamide and ethambutol between days 8 and 14. Results In the 40 mg·kg−1 cohort (n=15), 13 patients experienced a total of 36 adverse events during monotherapy, resulting in one treatment discontinuation. In the 50 mg·kg−1 cohort (n=17), all patients experienced adverse events during monotherapy, 93 in total; 11 patients withdrew or stopped study medication. Adverse events were mostly mild/moderate and tolerability rather than safety related, i.e. gastrointestinal disorders, pruritis, hyperbilirubinaemia and jaundice. There was a more than proportional increase in the rifampicin geometric mean area under the plasma concentration–time curve from time 0 to 12 h (AUC0–24 h) for 50 mg·kg−1 compared with 40 mg·kg−1; 571 (range 320–995) versus 387 (range 201–847) mg·L−1·h, while peak exposures saw proportional increases. Protein-unbound exposure after 50 mg·kg−1 (11% (range 8–17%)) was comparable with lower rifampicin doses. Rifampicin exposures and bilirubin concentrations were correlated (Spearman's ρ=0.670 on day 3, p<0.001). EBA increased considerably with dose, with the highest seen after 50 mg·kg−1: 14-day EBA −0.427 (95% CI −0.500– −0.355) log10CFU·mL−1·day−1. Conclusion Although associated with an increased bactericidal effect, the 50 mg·kg−1 dose was not well tolerated. Rifampicin at 40 mg·kg−1 was well tolerated and therefore selected for evaluation in a phase IIc treatment-shortening trial. While bactericidal activity continues to increase with dose, for the first time we identified dose-limiting intolerability for rifampicin dosed at 50 mg·kg−1; 40 mg·kg−1 seems the optimal tolerable dose for evaluation in TB treatment-shortening trialshttps://bit.ly/37dUIuB
Collapse
Affiliation(s)
- Lindsey H M Te Brake
- Dept of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Kim Narunsky
- UCT Lung Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Elin M Svensson
- Dept of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Dept of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Patrick P J Phillips
- UCSF Center for Tuberculosis, University of California San Francisco, San Francisco, CA, USA
| | - Stephen H Gillespie
- School of Medicine, Medical and Biological Sciences, University of St Andrews, St Andrews, UK
| | - Norbert Heinrich
- Division of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, Medical Center of the University of Munich, Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany
| | - Rodney Dawson
- UCT Lung Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Rob E Aarnoutse
- Dept of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | |
Collapse
|
7
|
Tátrai P, Krajcsi P. Prediction of Drug-Induced Hyperbilirubinemia by In Vitro Testing. Pharmaceutics 2020; 12:pharmaceutics12080755. [PMID: 32796590 PMCID: PMC7465333 DOI: 10.3390/pharmaceutics12080755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022] Open
Abstract
Bilirubin, the end product of heme catabolism, is produced continuously in the body and may reach toxic levels if accumulates in the serum and tissues; therefore, a highly efficient mechanism evolved for its disposition. Normally, unconjugated bilirubin enters hepatocytes through the uptake transporters organic anion transporting polypeptide (OATP) 1B1 and 1B3, undergoes glucuronidation by the Phase II enzyme UDP glucuronosyltransferase 1A1 (UGT1A1), and conjugated forms are excreted into the bile by the canalicular export pump multidrug resistance protein 2 (MRP2). Any remaining conjugated bilirubin is transported back to the blood by MRP3 and passed on for uptake and excretion by downstream hepatocytes or the kidney. The bile salt export pump BSEP as the main motor of bile flow is indirectly involved in bilirubin disposition. Genetic mutations and xenobiotics that interfere with this machinery may impede bilirubin disposition and cause hyperbilirubinemia. Several pharmaceutical compounds are known to cause hyperbilirubinemia via inhibition of OATP1Bs, UGT1A1, or BSEP. Herein we briefly review the in vitro prediction methods that serve to identify drugs with a potential to induce hyperbilirubinemia. In vitro assays can be deployed early in drug development and may help to minimize late-stage attrition. Based on current evidence, drugs that behave as mono- or multispecific inhibitors of OATP1B1, UGT1A1, and BSEP in vitro are at risk of causing clinically significant hyperbilirubinemia. By integrating inhibition data from in vitro assays, drug serum concentrations, and clinical reports of hyperbilirubinemia, predictor cut-off values have been established and are provisionally suggested in this review. Further validation of in vitro readouts to clinical outcomes is expected to enhance the predictive power of these assays.
Collapse
Affiliation(s)
- Péter Tátrai
- Solvo Biotechnology, Science Park, Building B1, 4-20 Irinyi József utca, H-1117 Budapest, Hungary;
| | - Péter Krajcsi
- Solvo Biotechnology, Science Park, Building B1, 4-20 Irinyi József utca, H-1117 Budapest, Hungary;
- Faculty of Health Sciences, Semmelweis University, H-1085 Budapest, Hungary
- Faculty of Information Technology and Bionics, Péter Pázmány Catholic University, H-1083 Budapest, Hungary
- Correspondence:
| |
Collapse
|
8
|
Physiologically-Based Pharmacokinetic (PBPK) Modeling of Buprenorphine in Adults, Children and Preterm Neonates. Pharmaceutics 2020; 12:pharmaceutics12060578. [PMID: 32585880 PMCID: PMC7355427 DOI: 10.3390/pharmaceutics12060578] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/18/2020] [Accepted: 06/21/2020] [Indexed: 12/15/2022] Open
Abstract
Buprenorphine plays a crucial role in the therapeutic management of pain in adults, adolescents and pediatric subpopulations. However, only few pharmacokinetic studies of buprenorphine in children, particularly neonates, are available as conducting clinical trials in this population is especially challenging. Physiologically-based pharmacokinetic (PBPK) modeling allows the prediction of drug exposure in pediatrics based on age-related physiological differences. The aim of this study was to predict the pharmacokinetics of buprenorphine in pediatrics with PBPK modeling. Moreover, the drug-drug interaction (DDI) potential of buprenorphine with CYP3A4 and P-glycoprotein perpetrator drugs should be elucidated. A PBPK model of buprenorphine and norbuprenorphine in adults has been developed and scaled to children and preterm neonates, accounting for age-related changes. One-hundred-percent of the predicted AUClast values in adults (geometric mean fold error (GMFE): 1.22), 90% of individual AUClast predictions in children (GMFE: 1.54) and 75% in preterm neonates (GMFE: 1.57) met the 2-fold acceptance criterion. Moreover, the adult model was used to simulate DDI scenarios with clarithromycin, itraconazole and rifampicin. We demonstrate the applicability of scaling adult PBPK models to pediatrics for the prediction of individual plasma profiles. The novel PBPK models could be helpful to further investigate buprenorphine pharmacokinetics in various populations, particularly pediatric subgroups.
Collapse
|
9
|
Singkham N, Avihingsanon A, Thammajaruk N, Ruxrungtham K, Bunupuradah T, Kiertiburanakul S, Chetchotisakd P, Burger DM, Emery S, Punyawudho B. Influence of CYP3A5 and SLCO1B1 polymorphisms on atazanavir/r concentrations in Thai HIV-infected patients. Pharmacogenomics 2020; 20:517-527. [PMID: 31124411 DOI: 10.2217/pgs-2018-0196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Aim: To evaluate the influence of genetic polymorphisms on plasma trough concentrations of atazanavir (ATV) and ritonavir (RTV). Patients & methods: The concentration-to-dose ratios were compared between different genotype groups of CYP3A5, ABCB1, SLCO1B1 and NR1I2 in 490 patients. Multiple regression analysis was used to examine the association between genetic and clinical factors and log-transformed concentration-to-dose ratio of ATV and RTV. Results: Higher concentrations of ATV and RTV were significantly associated with CYP3A5 6986 GG and SLCO1B1 521 TC or CC. Female patients had significantly higher ATV plasma concentration than male patients. Conclusion: Genetic polymorphisms and gender are factors affecting the variability of ATV and RTV concentrations in the Thai population. Thus, genetic testing is worth considering when atazanavir + low dose ritonavir is prescribed.
Collapse
Affiliation(s)
- Noppaket Singkham
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand.,PhD's Degree Program in Pharmacy, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Anchalee Avihingsanon
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand.,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Kiat Ruxrungtham
- HIV-NAT, Thai Red Cross AIDS Research Centre, Bangkok, Thailand.,Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Sasisopin Kiertiburanakul
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - David M Burger
- Department of Pharmacy, Radbound University Medical Center, Nijmegen, The Netherlands
| | - Sean Emery
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Baralee Punyawudho
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand.,Pharmacoepidemiology & Statistics Research Center (PESRC), Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
10
|
Sun R, Ying Y, Tang Z, Liu T, Shi F, Li H, Guo T, Huang S, Lai R. The Emerging Role of the SLCO1B3 Protein in Cancer Resistance. Protein Pept Lett 2020; 27:17-29. [PMID: 31556849 PMCID: PMC6978646 DOI: 10.2174/0929866526666190926154248] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/08/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Currently, chemotherapy is one of the mainstays of oncologic therapies. But the efficacy of chemotherapy is often limited by drug resistance and severe side effects. Consequently, it is becoming increasingly important to investigate the underlying mechanism and overcome the problem of anticancer chemotherapy resistance. The solute carrier organic anion transporter family member 1B3 (SLCO1B3), a functional transporter normally expressed in the liver, transports a variety of endogenous and exogenous compounds, including hormones and their conjugates as well as some anticancer drugs. The extrahepatic expression of SLCO1B3 has been detected in different cancer cell lines and cancer tissues. Recently, accumulating data indicates that the abnormal expression and function of SLCO1B3 are involved in resistance to anticancer drugs, such as taxanes, camptothecin and its analogs, SN-38, and Androgen Deprivation Therapy (ADT) in breast, prostate, lung, hepatic, and colorectal cancer, respectively. Thus, more investigations have been implemented to identify the potential SLCO1B3-related mechanisms of cancer drug resistance. In this review, we focus on the emerging roles of SLCO1B3 protein in the development of cancer chemotherapy resistance and briefly discuss the mechanisms of resistance. Elucidating the function of SLCO1B3 in chemoresistance may bring out novel therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Ruipu Sun
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China.,Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Zhimin Tang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Ting Liu
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Fuli Shi
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China
| | - Huixia Li
- Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Taichen Guo
- Nanchang Joint Program, Queen Mary University of London, London, United Kingdom
| | - Shibo Huang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University Medical College, Nanchang, China.,Department of Pharmacy, Medical College, Nanchang University, Nanchang 330006, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences / Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| |
Collapse
|
11
|
Vitek L, Bellarosa C, Tiribelli C. Induction of Mild Hyperbilirubinemia: Hype or Real Therapeutic Opportunity? Clin Pharmacol Ther 2019; 106:568-575. [PMID: 30588615 DOI: 10.1002/cpt.1341] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023]
Abstract
Observational epidemiological studies showed that mild hyperbilirubinemia has beneficial effects on the prevention of cardiovascular disease, type 2 diabetes mellitus, and metabolic syndrome. In mammals, bilirubin plays a major role as a potent antioxidant. Uridine 5'-diphospho-glucuronosyl transferase (UGT)1A1 variants coding for bilirubin UDP-glucuronosyl transferase resulting in mild hyperbilirubinemia (as in Gilbert syndrome (GS)) may confer a strong genetic advantage. Strategies to boost bioavailability of bilirubin or to mimic GS represent an attractive approach to prevent many oxidative stress and inflammation-mediated diseases. Even a tiny, micromolar increase in serum bilirubin concentrations substantially decreases the risk of oxidative stress-mediated diseases. There are several possible ways to achieve this, including lifestyle changes, changes in dietary patterns, regular physical activities, or use of chemical drug or of specific plant products either in the form of regular food items or nutraceuticals. Further basic and experimental research is required to fully uncover this promising therapeutic field.
Collapse
Affiliation(s)
- Libor Vitek
- Institute of Medical Biochemistry and Laboratory Diagnostics and 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cristina Bellarosa
- Fondazione Italiana Fegato ONLUS, AREA Science Park-Basovizza, Trieste, Italy
| | - Claudio Tiribelli
- Fondazione Italiana Fegato ONLUS, AREA Science Park-Basovizza, Trieste, Italy
| |
Collapse
|
12
|
Church RJ, Watkins PB. Serum biomarkers of drug-induced liver injury: Current status and future directions. J Dig Dis 2019; 20:2-10. [PMID: 30378260 DOI: 10.1111/1751-2980.12684] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
Drug-induced liver injury (DILI), which is caused by drugs and herbal or dietary supplements, remains a serious concern for drug developers, regulators, and clinicians; however, serum biomarkers utilized to detect and monitor DILI have not changed in decades and have limitations. Data-driven mathematical modeling that incorporates the release and clearance kinetics of traditional biomarkers has improved their use in the prediction of liver safety liabilities for new drug candidates. Several newer biomarkers have shown promise in terms of liver specificity, predicting the outcome of DILI events, and providing insight into its underlying mechanisms. For these new biomarkers to be qualified for regulatory acceptance, it will require their assessment in large numbers of patients who are receiving a wide range of compounds and who develop a broad spectrum of liver injuries. The ongoing and evolving international biomarker consortia should play a major role in this effort, which is likely to transform the assessment of liver safety in clinical trials and in the clinic.
Collapse
Affiliation(s)
- Rachel J Church
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Paul B Watkins
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina, USA.,Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Qosa H, Avaritt BR, Hartman NR, Volpe DA. In vitro UGT1A1 inhibition by tyrosine kinase inhibitors and association with drug-induced hyperbilirubinemia. Cancer Chemother Pharmacol 2018; 82:795-802. [PMID: 30105461 DOI: 10.1007/s00280-018-3665-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/03/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE Hyperbilirubinemia has been observed in patients treated with tyrosine kinase inhibitor (TKI) drugs. Therefore, it would be beneficial to understand whether there is a relationship between inhibition of uridine-5'-diphosphate glucuronosyltransferase (UGT) 1A1 activity and observed bilirubin elevations in TKI drug-treated patients. UGT1A1 is responsible for the glucuronidation of bilirubin which leads to its elimination in the bile. METHODS To examine this question, an in vitro glucuronidation assay was developed to determine the inhibitory effect of TKI drugs employing human liver microsomes (HLM) with varying UGT1A1 activity. Utilizing β-estradiol as the UGT1A1 probe substrate, 20 TKI drugs were evaluated at concentrations that represent clinical plasma levels. Adverse event reports were searched to generate an empirical Bayes geometric mean (EGBM) score for clinical hyperbilirubinemia with the TKI drugs. RESULTS Erlotinib, nilotinib, regorafenib, pazopanib, sorafenib and vemurafenib had IC50 values that were lower than their clinical steady-state Cmax concentrations. These TKI drugs had high incidences of hyperbilirubinemia and higher EBGM scores. The IC50 values and Cmax/IC50 ratios correlated well with EBGM scores for hyperbilirubinemia (P < 0.005). For the TKI drugs with higher incidence of hyperbilirubinemia in Gilbert syndrome patients, who have reduced UGT1A1 activity, six of eight had smaller ratios in the low UGT1A1 activity microsomes than the wild-type microsomes for drugs, indicating greater sensitivity to the drugs in this phenotype. CONCLUSIONS These results suggest that in vitro UGT1A1 inhibition assays have the potential to predict clinical hyperbilirubinemia.
Collapse
Affiliation(s)
- Hisham Qosa
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD, 20993, USA
| | - Brittany R Avaritt
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD, 20993, USA
- Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD, 20993, USA
| | - Neil R Hartman
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD, 20993, USA
| | - Donna A Volpe
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD, 20993, USA.
| |
Collapse
|
14
|
Zhang N, Shon J, Kim M, Yu C, Zhang L, Huang S, Lee L, Tran D, Li L. Role of CYP3A in Oral Contraceptives Clearance. Clin Transl Sci 2018; 11:251-260. [PMID: 28986954 PMCID: PMC5944580 DOI: 10.1111/cts.12499] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/26/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Nan Zhang
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
- Oak Ridge Institute for Science and Education (ORISE)TennesseeOak RidgeUSA
| | - Jihong Shon
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Myong‐Jin Kim
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Chongwoo Yu
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Lei Zhang
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Shiew‐Mei Huang
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - LaiMing Lee
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Doanh Tran
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| | - Li Li
- Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS)Center for Drug Evaluation and Research (CDER)US Food and Drug Administration (FDA)Silver SpringMarylandUSA
| |
Collapse
|
15
|
Chu X, Chan GH, Evers R. Identification of Endogenous Biomarkers to Predict the Propensity of Drug Candidates to Cause Hepatic or Renal Transporter-Mediated Drug-Drug Interactions. J Pharm Sci 2017; 106:2357-2367. [DOI: 10.1016/j.xphs.2017.04.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/18/2022]
|
16
|
Shebley M, Liu J, Kavetskaia O, Sydor J, de Morais SM, Fischer V, Nijsen MJMA, Bow DAJ. Mechanisms and Predictions of Drug-Drug Interactions of the Hepatitis C Virus Three Direct-Acting Antiviral Regimen: Paritaprevir/Ritonavir, Ombitasvir, and Dasabuvir. Drug Metab Dispos 2017; 45:755-764. [PMID: 28483778 DOI: 10.1124/dmd.116.074518] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/02/2017] [Indexed: 12/31/2022] Open
Abstract
To assess drug-drug interaction (DDI) potential for the three direct-acting antiviral (3D) regimen of ombitasvir, dasabuvir, and paritaprevir, in vitro studies profiled drug-metabolizing enzyme and transporter interactions. Using mechanistic static and dynamic models, DDI potential was predicted for CYP3A, CYP2C8, UDP-glucuronosyltransferase (UGT) 1A1, organic anion-transporting polypeptide (OATP) 1B1/1B3, breast cancer resistance protein (BCRP), and P-glycoprotein (P-gp). Perpetrator static model DDI predictions for metabolizing enzymes were within 2-fold of the clinical observations, but additional physiologically based pharmacokinetic modeling was necessary to achieve the same for drug transporters. When perpetrator interactions were assessed, ritonavir was responsible for the strong increase in exposure of sensitive CYP3A substrates, whereas paritaprevir (an OATP1B1/1B3 inhibitor) greatly increased the exposure of sensitive OATP1B1/1B3 substrates. The 3D regimen drugs are UGT1A1 inhibitors and are predicted to moderately increase plasma exposure of sensitive UGT1A1 substrates. Paritaprevir, ritonavir, and dasabuvir are BCRP inhibitors. Victim DDI predictions were qualitatively in line with the clinical observations. Plasma exposures of the 3D regimen were reduced by strong CYP3A inducers (paritaprevir and ritonavir; major CYP3A substrates) but were not affected by strong CYP3A4 inhibitors, since ritonavir (a CYP3A inhibitor) is already present in the regimen. Strong CYP2C8 inhibitors increased plasma exposure of dasabuvir (a major CYP2C8 substrate), OATP1B1/1B3 inhibitors increased plasma exposure of paritaprevir (an OATP1B1/1B3 substrate), and P-gp or BCRP inhibitors (all compounds are substrates of P-gp and/or BCRP) increased plasma exposure of the 3D regimen. Overall, the comprehensive mechanistic assessment of compound disposition along with mechanistic and PBPK approaches to predict victim and perpetrator DDI liability may enable better clinical management of nonstudied drug combinations with the 3D regimen.
Collapse
Affiliation(s)
- Mohamad Shebley
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| | - Jinrong Liu
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| | - Olga Kavetskaia
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| | - Jens Sydor
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| | - Sonia M de Morais
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| | - Volker Fischer
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| | - Marjoleen J M A Nijsen
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| | - Daniel A J Bow
- Drug Metabolism, Pharmacokinetics, and Bioanalysis (M.S., J.L., O.K., J.S., S.M.d.M., V.F., M.J.M.A.N., D.A.J.B.) and Clinical Pharmacology and Pharmacometrics (M.S.), AbbVie Inc., North Chicago, Illinois
| |
Collapse
|
17
|
Yang K, Battista C, Woodhead JL, Stahl SH, Mettetal JT, Watkins PB, Siler SQ, Howell BA. Systems pharmacology modeling of drug-induced hyperbilirubinemia: Differentiating hepatotoxicity and inhibition of enzymes/transporters. Clin Pharmacol Ther 2017; 101:501-509. [PMID: 28074467 PMCID: PMC5367379 DOI: 10.1002/cpt.619] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/31/2016] [Accepted: 01/04/2017] [Indexed: 12/20/2022]
Abstract
Elevations in serum bilirubin during drug treatment may indicate global liver dysfunction and a high risk of liver failure. However, drugs also can increase serum bilirubin in the absence of hepatic injury by inhibiting specific enzymes/transporters. We constructed a mechanistic model of bilirubin disposition based on known functional polymorphisms in bilirubin metabolism/transport. Using physiologically based pharmacokinetic (PBPK) model-predicted drug exposure and enzyme/transporter inhibition constants determined in vitro, our model correctly predicted indinavir-mediated hyperbilirubinemia in humans and rats. Nelfinavir was predicted not to cause hyperbilirubinemia, consistent with clinical observations. We next examined a new drug candidate that caused both elevations in serum bilirubin and biochemical evidence of liver injury in rats. Simulations suggest that bilirubin elevation primarily resulted from inhibition of transporters rather than global liver dysfunction. We conclude that mechanistic modeling of bilirubin can help elucidate underlying mechanisms of drug-induced hyperbilirubinemia, and thereby distinguish benign from clinically important elevations in serum bilirubin.
Collapse
Affiliation(s)
- K Yang
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| | - C Battista
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA.,University of North Carolina Institute for Drug Safety Sciences, The Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J L Woodhead
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| | - S H Stahl
- ADME Transporters, Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge, United Kingdom
| | - J T Mettetal
- Drug Safety and Metabolism, AstraZeneca R&D, Waltham, Massachusetts, USA
| | - P B Watkins
- University of North Carolina Institute for Drug Safety Sciences, The Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - S Q Siler
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| | - B A Howell
- DILIsym Services Inc, Research Triangle Park, North Carolina, USA
| |
Collapse
|
18
|
Lapham K, Novak J, Marroquin LD, Swiss R, Qin S, Strock CJ, Scialis R, Aleo MD, Schroeter T, Eng H, Rodrigues AD, Kalgutkar AS. Inhibition of Hepatobiliary Transport Activity by the Antibacterial Agent Fusidic Acid: Insights into Factors Contributing to Conjugated Hyperbilirubinemia/Cholestasis. Chem Res Toxicol 2016; 29:1778-1788. [DOI: 10.1021/acs.chemrestox.6b00262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | | | | | | | - Shuzhen Qin
- Biological
Screening and Assay Development, Cyprotex, Watertown, Massachusetts 02472, United States
| | - Christopher J. Strock
- Biological
Screening and Assay Development, Cyprotex, Watertown, Massachusetts 02472, United States
| | | | | | | | | | | | - Amit S. Kalgutkar
- Pharmacokinetics,
Dynamics, and Metabolism Department, Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
19
|
Kikuchi R, Peterkin VC, Chiou WJ, de Morais SM, Bow DAJ. Validation of a total IC50 method which enables in vitro assessment of transporter inhibition under semi-physiological conditions. Xenobiotica 2016; 47:825-832. [DOI: 10.1080/00498254.2016.1233372] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Ryota Kikuchi
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc, North Chicago, IL, USA
| | - Vincent C. Peterkin
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc, North Chicago, IL, USA
| | - William J. Chiou
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc, North Chicago, IL, USA
| | - Sonia M. de Morais
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc, North Chicago, IL, USA
| | - Daniel A. J. Bow
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc, North Chicago, IL, USA
| |
Collapse
|
20
|
Okubo H, Kitamura T, Ando H, Fukada H, Igusa Y, Kokubu S, Miyazaki A, Fujimura A, Shiina S, Watanabe S. Gadoxetic Acid-Enhanced MR Imaging Predicts Simeprevir-Induced Hyperbilirubinemia During Hepatitis C Virus Treatment: A Pilot Study. J Clin Pharmacol 2016; 57:369-375. [PMID: 27530761 DOI: 10.1002/jcph.811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/11/2016] [Indexed: 12/20/2022]
Abstract
Simeprevir is a substrate for organic anion-transporting polypeptides (OATPs) that transport bilirubin. Hyperbilirubinemia is an adverse event reported during treatment of chronic hepatitis C patients with simeprevir plus pegylated interferon and ribavirin. Because gadoxetic acid is also a substrate of OATPs, pretreatment gadoxetic acid-enhanced magnetic resonance imaging (MRI) may predict hyperbilirubinemia during treatment. This prospective study therefore evaluated 11 consecutive patients with chronic hepatitis C who underwent gadoxetic acid-enhanced MRI prior to treatment with simeprevir plus pegylated interferon and ribavirin for 12 weeks, followed by pegylated interferon and ribavirin for an additional 12 weeks. Their contrast enhancement index (CEI), an index of liver parenchymal enhancement during the hepatobiliary phase, was assessed before treatment. Plasma trough concentrations (Ctrough ) of simeprevir were determined 7 days after its administration, and serum bilirubin concentrations were measured throughout the course of treatment. Six patients (55%) developed hyperbilirubinemia (≥1.6 mg/dL) during treatment. Ctrough was significantly higher in patients with than without hyperbilirubinemia (P = .009), with a strong inverse relationship between CEI and Ctrough (r = -0.911, P < .001). CEI was significantly lower in patients with than without hyperbilirubinemia (P = .009), but there were no significant differences between the 2 groups in pretreatment serum albumin concentrations and FIB-4 index, an index of liver fibrosis. Hepatic enhancement with gadoxetic acid was related to Ctrough of simeprevir. Gadoxetic acid-enhanced magnetic resonance imaging may predict the development of hyperbilirubinemia during treatment of hepatitis C patients with simeprevir plus pegylated interferon and ribavirin.
Collapse
Affiliation(s)
- Hironao Okubo
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Tsuneo Kitamura
- Department of Gastroenterology, Juntendo University Urayasu Hospital, Tokyo, Japan
| | - Hitoshi Ando
- Division of Clinical Pharmacology, Department of Pharmacology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Hiroo Fukada
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Yuki Igusa
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Shigehiro Kokubu
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Akihisa Miyazaki
- Department of Gastroenterology, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Akio Fujimura
- Division of Clinical Pharmacology, Department of Pharmacology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shuichiro Shiina
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Sumio Watanabe
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Sahota T, Danhof M, Della Pasqua O. Pharmacology-based toxicity assessment: towards quantitative risk prediction in humans. Mutagenesis 2016; 31:359-74. [PMID: 26970519 DOI: 10.1093/mutage/gev081] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite ongoing efforts to better understand the mechanisms underlying safety and toxicity, ~30% of the attrition in drug discovery and development is still due to safety concerns. Changes in current practice regarding the assessment of safety and toxicity are required to reduce late stage attrition and enable effective development of novel medicines. This review focuses on the implications of empirical evidence generation for the evaluation of safety and toxicity during drug development. A shift in paradigm is needed to (i) ensure that pharmacological concepts are incorporated into the evaluation of safety and toxicity; (ii) facilitate the integration of historical evidence and thereby the translation of findings across species as well as between in vitro and in vivo experiments and (iii) promote the use of experimental protocols tailored to address specific safety and toxicity questions. Based on historical examples, we highlight the challenges for the early characterisation of the safety profile of a new molecule and discuss how model-based methodologies can be applied for the design and analysis of experimental protocols. Issues relative to the scientific rationale are categorised and presented as a hierarchical tree describing the decision-making process. Focus is given to four different areas, namely, optimisation, translation, analytical construct and decision criteria. From a methodological perspective, the relevance of quantitative methods for estimation and extrapolation of risk from toxicology and safety pharmacology experimental protocols, such as points of departure and potency, is discussed in light of advancements in population and Bayesian modelling techniques (e.g. non-linear mixed effects modelling). Their use in the evaluation of pharmacokinetics (PK) and pharmacokinetic-pharmacodynamic relationships (PKPD) has enabled great insight into the dose rationale for medicines in humans, both in terms of efficacy and adverse events. Comparable benefits can be anticipated for the assessment of safety and toxicity profile of novel molecules.
Collapse
Affiliation(s)
- Tarjinder Sahota
- Division of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
| | - Meindert Danhof
- Division of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands
| | - Oscar Della Pasqua
- Division of Pharmacology, Leiden Academic Centre for Drug Research, University of Leiden, Leiden, The Netherlands, Clinical Pharmacology, Modelling and Simulation, GlaxoSmithKline, Stockley Park West, Uxbridge, UK, Clinical Pharmacology and Therapeutics, University College London, London, UK
| |
Collapse
|
22
|
Eng H, Scialis RJ, Rotter CJ, Lin J, Lazzaro S, Varma MV, Di L, Feng B, West M, Kalgutkar AS. The Antimicrobial Agent Fusidic Acid Inhibits Organic Anion Transporting Polypeptide-Mediated Hepatic Clearance and May Potentiate Statin-Induced Myopathy. Drug Metab Dispos 2016; 44:692-9. [DOI: 10.1124/dmd.115.067447] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/12/2016] [Indexed: 12/19/2022] Open
|
23
|
Li X, Zhong K, Guo Z, Zhong D, Chen X. Fasiglifam (TAK-875) Inhibits Hepatobiliary Transporters: A Possible Factor Contributing to Fasiglifam-Induced Liver Injury. Drug Metab Dispos 2015; 43:1751-9. [PMID: 26276582 DOI: 10.1124/dmd.115.064121] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/12/2015] [Indexed: 12/20/2022] Open
Abstract
Fasiglifam (TAK-875), a selective G-protein-coupled receptor 40 agonist, was developed for the treatment of type 2 diabetes mellitus; however, its development was terminated in phase III clinical trials because of liver safety concerns. Our preliminary study indicated that intravenous administration of 100 mg/kg of TAK-875 increased the serum total bile acid concentration by 3 to 4 times and total bilirubin levels by 1.5 to 2.6 times in rats. In the present study, we examined the inhibitory effects of TAK-875 on hepatobiliary transporters to explore the mechanisms underlying its hepatotoxicity. TAK-875 decreased the biliary excretion index and the in vitro biliary clearance of d₈-taurocholic acid in sandwich-cultured rat hepatocytes, suggesting that TAK-875 impaired biliary excretion of bile acids, possibly by inhibiting bile salt export pump (Bsep). TAK-875 inhibited the efflux transporter multidrug resistance-associated protein 2 (Mrp2) in rat hepatocytes using 5 (and 6)-carboxy-2',7'-dichlorofluorescein as a substrate. Inhibition of MRP2 was further confirmed by reduced transport of vinblastine in Madin-Darby canine kidney cells overexpressing MRP2 with IC₅₀ values of 2.41 μM. TAK-875 also inhibited the major bile acid uptake transporter Na(+)/taurocholate cotransporting polypeptide (Ntcp), which transports d₈-taurocholic acid into rat hepatocytes, with an IC₅₀ value of 10.9 μM. TAK-875 significantly inhibited atorvastatin uptake in organic anion transporter protein (OATP) 1B1 and OATP1B3 cells with IC₅₀ values of 2.28 and 3.98 μM, respectively. These results indicate that TAK-875 inhibited the efflux transporter MRP2/Mrp2 and uptake transporters Ntcp and OATP/Oatp, which may affect bile acid and bilirubin homeostasis, resulting in hyperbilirubinemia and cholestatic hepatotoxicity.
Collapse
Affiliation(s)
- Xiuli Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Kan Zhong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zitao Guo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Dafang Zhong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoyan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
24
|
Chu X, Shih SJ, Shaw R, Hentze H, Chan GH, Owens K, Wang S, Cai X, Newton D, Castro-Perez J, Salituro G, Palamanda J, Fernandis A, Ng CK, Liaw A, Savage MJ, Evers R. Evaluation of cynomolgus monkeys for the identification of endogenous biomarkers for hepatic transporter inhibition and as a translatable model to predict pharmacokinetic interactions with statins in humans. Drug Metab Dispos 2015; 43:851-63. [PMID: 25813937 DOI: 10.1124/dmd.115.063347] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/26/2015] [Indexed: 12/31/2022] Open
Abstract
Inhibition of hepatic transporters such as organic anion transporting polypeptides (OATPs) 1B can cause drug-drug interactions (DDIs). Determining the impact of perpetrator drugs on the plasma exposure of endogenous substrates for OATP1B could be valuable to assess the risk for DDIs early in drug development. As OATP1B orthologs are well conserved between human and monkey, we assessed in cynomolgus monkeys the endogenous OATP1B substrates that are potentially suitable to assess DDI risk in humans. The effect of rifampin (RIF), a potent inhibitor for OATP1B, on plasma exposure of endogenous substrates of hepatic transporters was measured. From the 18 biomarkers tested, RIF (18 mg/kg, oral) caused significant elevation of plasma unconjugated and conjugated bilirubin, which may be attributed to inhibition of cOATP1B1 and cOATP1B3 based on in vitro to in vivo extrapolation analysis. To further evaluate whether cynomolgus monkeys are a suitable translational model to study OATP1B-mediated DDIs, we determined the inhibitory effect of RIF on in vitro transport and pharmacokinetics of rosuvastatin (RSV) and atorvastatin (ATV). RIF strongly inhibited the uptake of RSV and ATV by cOATP1B1 and cOATP1B3 in vitro. In agreement with clinical observations, RIF (18 mg/kg, oral) significantly decreased plasma clearance and increased the area under the plasma concentration curve (AUC) of intravenously administered RSV by 2.8- and 2.7-fold, and increased the AUC and maximum plasma concentration of orally administered RSV by 6- and 10.3-fold, respectively. In contrast to clinical findings, RIF did not significantly increase plasma exposure of either intravenous or orally administered ATV, indicating species differences in the rate-limiting elimination pathways.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Shian-Jiun Shih
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Rachel Shaw
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Hannes Hentze
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Grace H Chan
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Karen Owens
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Shubing Wang
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Xiaoxin Cai
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Deborah Newton
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Jose Castro-Perez
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Gino Salituro
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Jairam Palamanda
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Aaron Fernandis
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Choon Keow Ng
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Andy Liaw
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Mary J Savage
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| | - Raymond Evers
- Merck Sharp & Dohme Corporation, Kenilworth, New Jersey (X.C., G.H.C., K.O., S.W., X.C., D.N., J.C.P., G.S., J.P., A.L., M.J.S., R.E.); Translational Medicine Research Centre, Singapore (S.J.S., R.S., H.H., A.F., C.K.N.)
| |
Collapse
|
25
|
Watanabe T, Miyake M, Shimizu T, Kamezawa M, Masutomi N, Shimura T, Ohashi R. Utility of bilirubins and bile acids as endogenous biomarkers for the inhibition of hepatic transporters. Drug Metab Dispos 2015; 43:459-66. [PMID: 25581390 DOI: 10.1124/dmd.114.061051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It is useful to identify endogenous substrates for the evaluation of drug-drug interactions via transporters. In this study, we investigated the utility of bilirubins, substrates of OATPs and MRP2, and bile acids and substrates of NTCP and BSEP, as biomarkers for the inhibition of transporters. In rats administered 20 and 80 mg/kg rifampicin, the plasma levels of bilirubin glucuronides were elevated, gradually decreased, and almost returned to the baseline level at 24 hours after administration without an elevation of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). This result indicates the transient inhibition of rOatps and/or rMrp2. Although the correlation between free plasma concentrations and IC50 values of rOatps depended on the substrates used in the in vitro studies, the inhibition of rOatps by rifampicin was confirmed in the in vivo study using valsartan as a substrate of rOatps. In rats administered 10 and 30 mg/kg cyclosporin A, the plasma levels of bile acids were elevated and persisted for up to 24 hours after administration without an elevation of ALT and AST. This result indicates the continuous inhibition of rNtcp and/or rBsep, although there were differences between the free plasma or liver concentrations and IC50 values of rNtcp or rBsep, respectively. This study suggests that the monitoring of bilirubins and bile acids in plasma is useful in evaluating the inhibitory potential of their corresponding transporters.
Collapse
Affiliation(s)
- Tomoko Watanabe
- DMPK Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan (T.W., M.K., Ta.S., R.O.); and Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan (M.M., To.S., N.M.)
| | - Manami Miyake
- DMPK Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan (T.W., M.K., Ta.S., R.O.); and Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan (M.M., To.S., N.M.)
| | - Toshinobu Shimizu
- DMPK Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan (T.W., M.K., Ta.S., R.O.); and Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan (M.M., To.S., N.M.)
| | - Miho Kamezawa
- DMPK Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan (T.W., M.K., Ta.S., R.O.); and Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan (M.M., To.S., N.M.)
| | - Naoya Masutomi
- DMPK Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan (T.W., M.K., Ta.S., R.O.); and Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan (M.M., To.S., N.M.)
| | - Takesada Shimura
- DMPK Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan (T.W., M.K., Ta.S., R.O.); and Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan (M.M., To.S., N.M.)
| | - Rikiya Ohashi
- DMPK Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan (T.W., M.K., Ta.S., R.O.); and Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan (M.M., To.S., N.M.)
| |
Collapse
|
26
|
Sane RS, Steinmann GG, Huang Q, Li Y, Podila L, Mease K, Olson S, Taub ME, Stern JO, Nehmiz G, Böcher WO, Asselah T, Tweedie D. Mechanisms underlying benign and reversible unconjugated hyperbilirubinemia observed with faldaprevir administration in hepatitis C virus patients. J Pharmacol Exp Ther 2014; 351:403-12. [PMID: 25204339 DOI: 10.1124/jpet.114.218081] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Faldaprevir, an investigational agent for hepatitis C virus treatment, is well tolerated but associated with rapidly reversible, dose-dependent, clinically benign, unconjugated hyperbilirubinemia. Multidisciplinary preclinical and clinical studies were used to characterize mechanisms underlying this hyperbilirubinemia. In vitro, faldaprevir inhibited key processes involved in bilirubin clearance: UDP glucuronosyltransferase (UGT) 1A1 (UGT1A1) (IC50 0.45 µM), which conjugates bilirubin, and hepatic uptake and efflux transporters, organic anion-transporting polypeptide (OATP) 1B1 (IC50 0.57 µM), OATP1B3 (IC50 0.18 µM), and multidrug resistance-associated protein (MRP) 2 (IC50 6.2 µM), which transport bilirubin and its conjugates. In rat and human hepatocytes, uptake and biliary excretion of [(3)H]bilirubin and/or its glucuronides decreased on coincubation with faldaprevir. In monkeys, faldaprevir (≥20 mg/kg per day) caused reversible unconjugated hyperbilirubinemia, without hemolysis or hepatotoxicity. In clinical studies, faldaprevir-mediated hyperbilirubinemia was predominantly unconjugated, and levels of unconjugated bilirubin correlated with the UGT1A1*28 genotype. The reversible and dose-dependent nature of the clinical hyperbilirubinemia was consistent with competitive inhibition of bilirubin clearance by faldaprevir, and was not associated with liver toxicity or other adverse events. Overall, the reversible, unconjugated hyperbilirubinemia associated with faldaprevir may predominantly result from inhibition of bilirubin conjugation by UGT1A1, with inhibition of hepatic uptake of bilirubin also potentially playing a role. Since OATP1B1/1B3 are known to be involved in hepatic uptake of circulating bilirubin glucuronides, inhibition of OATP1B1/1B3 and MRP2 may underlie isolated increases in conjugated bilirubin. As such, faldaprevir-mediated hyperbilirubinemia is not associated with any liver injury or toxicity, and is considered to result from decreased bilirubin elimination due to a drug-bilirubin interaction.
Collapse
Affiliation(s)
- Rucha S Sane
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Gerhard G Steinmann
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Qihong Huang
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Yongmei Li
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Lalitha Podila
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Kirsten Mease
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Stephen Olson
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Mitchell E Taub
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Jerry O Stern
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Gerhard Nehmiz
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Wulf O Böcher
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Tarik Asselah
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| | - Donald Tweedie
- Boehringer Ingelheim Pharma Inc., Ridgefield, Connecticut (R.S.S., Q.H., Y.L., L.P., K.M., S.O., M.E.T., J.O.S., D.T.); Boehringer Ingelheim Pharma GmbH&Co. KG, Biberach, Germany (G.G.S., G.N.); Boehringer Ingelheim Pharma GmbH, Ingelheim, Germany (W.O.B.); and Hôpital Beaujon and INSERM UMR773, Université Denis Diderot Paris 7, Paris, France (T.A.)
| |
Collapse
|
27
|
Bortolussi G, Zentillin L, Vaníkova J, Bockor L, Bellarosa C, Mancarella A, Vianello E, Tiribelli C, Giacca M, Vitek L, Muro AF. Life-long correction of hyperbilirubinemia with a neonatal liver-specific AAV-mediated gene transfer in a lethal mouse model of Crigler-Najjar Syndrome. Hum Gene Ther 2014; 25:844-55. [PMID: 25072305 PMCID: PMC4175423 DOI: 10.1089/hum.2013.233] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 07/21/2014] [Indexed: 12/21/2022] Open
Abstract
Null mutations in the UGT1A1 gene result in Crigler-Najjar syndrome type I (CNSI), characterized by severe hyperbilirubinemia and constant risk of developing neurological damage. Phototherapy treatment lowers plasma bilirubin levels, but its efficacy is limited and liver transplantation is required. To find alternative therapies, we applied AAV liver-specific gene therapy to a lethal mouse model of CNSI. We demonstrated that a single neonatal hUGT1A1 gene transfer was successful and the therapeutic effect lasted up to 17 months postinjection. The therapeutic effect was mediated by the presence of transcriptionally active double-stranded episomes. We also compared the efficacy of two different gene therapy approaches: liver versus skeletal muscle transgene expression. We observed that 5-8% of normal liver expression and activity levels were sufficient to significantly reduce bilirubin levels and maintain lifelong low plasma bilirubin concentration (3.1±1.5 mg/dl). In contrast, skeletal muscle was not able to efficiently lower bilirubin (6.4±2.0 mg/dl), despite 20-30% of hUgt1a1 expression levels, compared with normal liver. We propose that this remarkable difference in gene therapy efficacy could be related to the absence of the Mrp2 and Mrp3 transporters of conjugated bilirubin in muscle. Taken together, our data support the concept that liver is the best organ for efficient and long-term CNSI gene therapy, and suggest that the use of extra-hepatic tissues should be coupled to the presence of bilirubin transporters.
Collapse
Affiliation(s)
- Giulia Bortolussi
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Lorena Zentillin
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Jana Vaníkova
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
| | - Luka Bockor
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Cristina Bellarosa
- Centro Studi Fegato, Fondazione Italiana Fegato, AREA Science Park, Campus Basovizza, 34149 Trieste, Italy
| | - Antonio Mancarella
- Centro Studi Fegato, Fondazione Italiana Fegato, AREA Science Park, Campus Basovizza, 34149 Trieste, Italy
| | - Eleonora Vianello
- Centro Studi Fegato, Fondazione Italiana Fegato, AREA Science Park, Campus Basovizza, 34149 Trieste, Italy
| | - Claudio Tiribelli
- Centro Studi Fegato, Fondazione Italiana Fegato, AREA Science Park, Campus Basovizza, 34149 Trieste, Italy
- Department of Medical Science, University of Trieste, 34128 Trieste, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Medicine, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
- Fourth Department of Internal Medicine, First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic
| | - Andrés F. Muro
- International Centre for Genetic Engineering and Biotechnology, 34149 Trieste, Italy
| |
Collapse
|
28
|
Different interaction profiles of direct-acting anti-hepatitis C virus agents with human organic anion transporting polypeptides. Antimicrob Agents Chemother 2014; 58:4555-64. [PMID: 24867984 DOI: 10.1128/aac.02724-14] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Simeprevir (SMV), asunaprevir (ASV), daclatasvir (DCV), and sofosbuvir (SFV), which are newly developed direct-acting antiviral agents (DAAs) against hepatitis C virus (HCV) infection, are among the key components of anti-HCV regimens. Preclinical studies have identified inhibitory properties for some of these DAAs against organic anion transporting polypeptide 1B (OATP1B) functions. However, their details remain mostly uncharacterized. Because OATP1B1 and OATP1B3 play determinant roles in the pharmacokinetics of various drugs via their uptake into human hepatocytes, it is plausible that the inhibition of these OATP1Bs by a DAA would create a potential risk of drug-drug interaction, which has been an emerging concern in anti-HCV therapy. Accordingly, in the present study, we intended to clarify the inhibitory characteristics of newly developed DAAs toward OATP1B1 and -1B3 functions. The results of our coincubation inhibition assays have shown that all tested DAAs could inhibit OATP1B1 functions and that SMV, ASV, and DCV (to a lesser extent), but not SFV, exhibited long-lasting preincubation inhibitory effects on OATP1B1 functions. It was also found that the preincubation inhibitory effects of SMV and ASV could augment their coincubation inhibition potency. Furthermore, significant, but differential, inhibitory effects of the DAAs on the OATP1B3 function were identified. To summarize, our results clearly show that the newly developed DAAs are newly identified OATP1B1 and OATP1B3 inhibitors with distinctive interaction properties. It is believed that these inhibition profiles will provide essential information to all concerned parties with respect to the clinical significance of DAA-mediated inhibition of OATP1Bs in anti-HCV therapy.
Collapse
|