Pharmacologic Treatment of Transplant Recipients Infected With SARS-CoV-2: Considerations Regarding Therapeutic Drug Monitoring and Drug–Drug Interactions

Therapeutic Drug Monitoring - Tập 42 Số 3 - Trang 360-368 - 2020
Laure Elens1,2, Loralie J. Langman3, Dennis A. Hesselink4,5, Stein Bergan6, Dirk Jan A. R. Moes7, Mariadelfina Molinaro8, Raman Venkataramanan9, Florian Lemaître10,11
1Institut de Recherche Expérimentale et Clinique (IREC), Louvain Center for Toxicology and Applied Pharmacology (LTAP), Université catholique de Louvain (UCLouvain), Brussels, Belgium;
2Louvain Drug Research Institute (LDRI), Integrated Pharmacometrics, Pharmacogenomics and Pharmacokinetics (PMGK), Université catholique de Louvain (UCLouvain), Brussels, Belgium;
3Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
4Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
5Rotterdam Transplant Group;
6Department of Pharmacology, Oslo University Hospital, Oslo, Norway
7Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
8Clinical and Experimental Pharmacokinetics Lab, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy;
9School of Pharmacy and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania;
10INSERM, Centre d'Investigation Clinique CIC 1414, Rennes, France
11Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)—UMR_S 1085, Rennes, France; and

Tóm tắt

Background: COVID-19 is a novel infectious disease caused by the severe acute respiratory distress (SARS)-coronavirus-2 (SARS-CoV-2). Several therapeutic options are currently emerging but none with universal consensus or proven efficacy. Solid organ transplant recipients are perceived to be at increased risk of severe COVID-19 because of their immunosuppressed conditions due to chronic use of immunosuppressive drugs (ISDs). It is therefore likely that solid organ transplant recipients will be treated with these experimental antivirals. Methods: This article is not intended to provide a systematic literature review on investigational treatments tested against COVID-19; rather, the authors aim to provide recommendations for therapeutic drug monitoring of ISDs in transplant recipients infected with SARS-CoV-2 based on a review of existing data in the literature. Results: Management of drug–drug interactions between investigational anti-SARS-CoV-2 drugs and immunosuppressants is a complex task for the clinician. Adequate immunosuppression is necessary to prevent graft rejection while, if critically ill, the patient may benefit from pharmacotherapeutic interventions directed at limiting SARS-CoV-2 viral replication. Maintaining ISD concentrations within the desired therapeutic range requires a highly individualized approach that is complicated by the pandemic context and lack of hindsight. Conclusions: With this article, the authors inform the clinician about the potential interactions of experimental COVID-19 treatments with ISDs used in transplantation. Recommendations regarding therapeutic drug monitoring and dose adjustments in the context of COVID-19 are provided.

Từ khóa


Tài liệu tham khảo

Dong, 2020, Discovering drugs to treat coronavirus disease 2019 (COVID-19), Drug Discov Ther., 14, 58, 10.5582/ddt.2020.01012

Li, 2020, Updated approaches against SARS-CoV-2, Antimicrob Agents Chemother.

Pedersen, 2020, SARS-CoV-2: a storm is raging, J Clin Invest., 10.1172/JCI137647

Suzuki, 2019, Factors involved in phenoconversion of CYP3A using 4beta-hydroxycholesterol in stable kidney transplant recipients, Pharmacol Rep., 71, 276, 10.1016/j.pharep.2018.12.007

Elens, 2012, Pharmacogenetics in kidney transplantation: recent updates and potential clinical applications, Mol Diagn Ther., 16, 331, 10.1007/s40291-012-0012-5

Benjanuwattra, 2020, Mycophenolic acid and its pharmacokinetic drug-drug interactions in humans: review of the evidence and clinical implications, J Clin Pharmacol., 60, 295, 10.1002/jcph.1565

Hesselink, 2005, Cyclosporine interacts with mycophenolic acid by inhibiting the multidrug resistance-associated protein 2, Am J Transpl., 5, 987, 10.1046/j.1600-6143.2005.00779.x

Kumar, 2007, Antimalarial drugs inhibiting hemozoin (beta-hematin) formation: a mechanistic update, Life Sci., 80, 813, 10.1016/j.lfs.2006.11.008

Liu, 2020, Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro, Cell Discov., 6, 16, 10.1038/s41421-020-0156-0

Yao, 2020, In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Clin Infect Dis., 10.1093/cid/ciaa237

Gautret, 2020, Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial, Int J Antimicrob Agents, 105949, 10.1016/j.ijantimicag.2020.105949

Furst, 1996, Pharmacokinetics of hydroxychloroquine and chloroquine during treatment of rheumatic diseases, Lupus., 5, S11, 10.1177/0961203396005001041

Kim, 2003, Cytochrome P450 2C8 and CYP3A4/5 are involved in chloroquine metabolism in human liver microsomes, Arch Pharm Res., 26, 631, 10.1007/BF02976712

Lim, 2009, Pharmacokinetics of hydroxychloroquine and its clinical implications in chemoprophylaxis against malaria caused by Plasmodium vivax, Antimicrob Agents Chemother., 53, 1468, 10.1128/AAC.00339-08

Ducharme, 1996, Clinical pharmacokinetics and metabolism of chloroquine. Focus on recent advancements, Clin Pharmacokinet., 31, 257, 10.2165/00003088-199631040-00003

Schrezenmeier, 2020, Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology, Nat Rev Rheumatol., 16, 155, 10.1038/s41584-020-0372-x

Chen, 2006, Chronic hydroxychloroquine use associated with QT prolongation and refractory ventricular arrhythmia, Clin Toxicol., 44, 173, 10.1080/15563650500514558

Ikitimur, 2015, Long-Term impact of different immunosuppressive drugs on QT and PR intervals in renal transplant patients, Ann Noninvasive Electrocardiol., 20, 426, 10.1111/anec.12225

White, 2007, Cardiotoxicity of antimalarial drugs, Lancet Infect Dis., 7, 549, 10.1016/S1473-3099(07)70187-1

Urva, 2013, A phase I study evaluating the effect of everolimus on the pharmacokinetics of midazolam in healthy subjects, J Clin Pharmacol., 53, 444, 10.1002/jcph.7

Finielz, 1993, Interaction between cyclosporin and chloroquine, Nephron., 65, 333, 10.1159/000187506

Nampoory, 1992, Drug interaction of chloroquine with ciclosporin, Nephron., 62, 108, 10.1159/000187007

Tan, 1991, Drug interaction between cyclosporine A and quinine in a renal transplant patient with malaria, Singap Med J., 32, 189

Sarzi-Puttini, 2005, An open, randomized comparison study of cyclosporine A, cyclosporine A + methotrexate and cyclosporine A + hydroxychloroquine in the treatment of early severe rheumatoid arthritis, Rheumatol Int., 25, 15, 10.1007/s00296-003-0384-2

Salaffi, 1996, Combination therapy of cyclosporine A with methotrexate or hydroxychloroquine in refractory rheumatoid arthritis, Scand J Rheumatol., 25, 16, 10.3109/03009749609082662

Soichot, 2014, Development, validation and clinical application of a LC-MS/MS method for the simultaneous quantification of hydroxychloroquine and its active metabolites in human whole blood, J Pharm Biomed Anal., 100, 131, 10.1016/j.jpba.2014.07.009

Kaewkhao, 2019, High sensitivity methods to quantify chloroquine and its metabolite in human blood samples using LC-MS/MS, Bioanalysis., 11, 333, 10.4155/bio-2018-0202

Morita, 2016, Population pharmacokinetics of hydroxychloroquine in Japanese patients with cutaneous or systemic lupus erythematosus, Ther Drug Monit., 38, 259, 10.1097/FTD.0000000000000261

Chen, 2004, In vitro susceptibility of 10 clinical isolates of SARS coronavirus to selected antiviral compounds, J Clin Virol., 31, 69, 10.1016/j.jcv.2004.03.003

Zhang, 2004, Old drugs as lead compounds for a new disease? Binding analysis of SARS coronavirus main proteinase with HIV, psychotic and parasite drugs, Bioorg Med Chem., 12, 2517, 10.1016/j.bmc.2004.03.035

Hurst, 2000, Lopinavir, Drugs., 60, 1371, 10.2165/00003495-200060060-00009

de Wilde, 2014, Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture, Antimicrob Agents Chemother., 58, 4875, 10.1128/AAC.03011-14

Ortega, 2020, Unrevealing sequence and structural features of novel coronavirus using in silico approaches: the main protease as molecular target, EXCLI J., 19, 400

Cao, 2020, A trial of lopinavir-ritonavir in adults hospitalized with severe covid-19, N Engl J Med., 10.1056/NEJMoa2001282

Konig, 2010, Impact of drug transporters on cellular resistance towards saquinavir and darunavir, J Antimicrob Chemother., 65, 2319, 10.1093/jac/dkq324

Svard, 2010, Nuclear receptor-mediated induction of CYP450 by antiretrovirals: functional consequences of NR1I2 (PXR) polymorphisms and differential prevalence in whites and sub-Saharan Africans, J Acquir Immune Defic Syndr., 55, 536, 10.1097/QAI.0b013e3181f52f0c

Holmstock, 2018, MRP2 inhibition by HIV protease inhibitors in rat and human hepatocytes: a quantitative confocal microscopy study, Drug Metab Dispos., 46, 697, 10.1124/dmd.117.079467

Qazi, 2002, Lopinavir/ritonavir (ABT-378/r), Expert Opin Pharmacother., 3, 315, 10.1517/14656566.3.3.315

Lepist, 2012, Cobicistat boosts the intestinal absorption of transport substrates, including HIV protease inhibitors and GS-7340, in vitro, Antimicrob Agents Chemother., 56, 5409, 10.1128/AAC.01089-12

Hsu, 1998, Ritonavir. Clinical pharmacokinetics and interactions with other anti-HIV agents, Clin Pharmacokinet., 35, 275, 10.2165/00003088-199835040-00002

Larson, 2014, Pharmacokinetic enhancers in HIV therapeutics, Clin Pharmacokinet., 53, 865, 10.1007/s40262-014-0167-9

Hu, 2014, Transcriptional regulation of human UDP-glucuronosyltransferase genes, Drug Metab Rev., 46, 421, 10.3109/03602532.2014.973037

Cattaneo, 2019, Pharmacokinetic drug evaluation of ritonavir (versus cobicistat) as adjunctive therapy in the treatment of HIV, Expert Opin Drug Metab Toxicol., 15, 927, 10.1080/17425255.2019.1685495

van Maarseveen, 2012, Drug-drug interactions between antiretroviral and immunosuppressive agents in HIV-infected patients after solid organ transplantation: a review, AIDS Patient Care STDS., 26, 568, 10.1089/apc.2012.0169

Marfo, 2009, Antiretroviral and immunosuppressive drug-drug interactions in human immunodeficiency virus-infected liver and kidney transplant recipients, Transpl Proc., 41, 3796, 10.1016/j.transproceed.2009.06.186

Frassetto, 2007, Immunosuppressant pharmacokinetics and dosing modifications in HIV-1 infected liver and kidney transplant recipients, Am J Transpl., 7, 2816, 10.1111/j.1600-6143.2007.02007.x

Jain, 2003, Effect of coadministered lopinavir and ritonavir (Kaletra) on tacrolimus blood concentration in liver transplantation patients, Liver Transpl., 9, 954, 10.1053/jlts.2003.50171

Bickel, 2010, Daily dosing of tacrolimus in patients treated with HIV-1 therapy containing a ritonavir-boosted protease inhibitor or raltegravir, J Antimicrob Chemother., 65, 999, 10.1093/jac/dkq054

Jain, 2002, Nelfinavir, a protease inhibitor, increases sirolimus levels in a liver transplantation patient: a case report, Liver Transpl., 8, 838, 10.1053/jlts.2002.34921

Krown, 2012, Rapamycin with antiretroviral therapy in AIDS-associated Kaposi sarcoma: an AIDS Malignancy Consortium Study, J Acquir Immune Defic Syndr., 59, 447, 10.1097/QAI.0b013e31823e7884

Frassetto, 2014, Best single time point correlations with AUC for cyclosporine and tacrolimus in HIV-infected kidney and liver transplant recipients, Transplantation., 97, 702, 10.1097/01.TP.0000441097.30094.31

Frassetto, 2005, Cyclosporine pharmacokinetics and dosing modifications in human immunodeficiency virus-infected liver and kidney transplant recipients, Transplantation., 80, 13, 10.1097/01.TP.0000165111.09687.4E

Brunet, 2019, Therapeutic drug monitoring of tacrolimus-personalized therapy: second consensus report, Ther Drug Monit., 41, 261, 10.1097/FTD.0000000000000640

Benkali, 2010, Population pharmacokinetics and Bayesian estimation of tacrolimus exposure in renal transplant recipients on a new once-daily formulation, Clin Pharmacokinet., 49, 683, 10.2165/11535950-000000000-00000

Vethe, 2019, Tacrolimus can Be reliably measured with volumetric absorptive capillary microsampling throughout the dose interval in renal transplant recipients, Ther Drug Monit., 41, 607, 10.1097/FTD.0000000000000655

Leger, 2002, Maximum a posteriori Bayesian estimation of oral cyclosporin pharmacokinetics in patients with stable renal transplants, Clin Pharmacokinet., 41, 71, 10.2165/00003088-200241010-00006

Monchaud, 2003, Limited sampling strategies using Bayesian estimation or multilinear regression for cyclosporin AUC(0-12) monitoring in cardiac transplant recipients over the first year post-transplantation, Eur J Clin Pharmacol., 58, 813, 10.1007/s00228-003-0559-5

Velghe, 2018, Dried blood spots in therapeutic drug monitoring and toxicology, Expert Opin Drug Metab Toxicol., 14, 1, 10.1080/17425255.2018.1414181

Scholten, 2005, AUC-guided dosing of tacrolimus prevents progressive systemic overexposure in renal transplant recipients, Kidney Int., 67, 2440, 10.1111/j.1523-1755.2005.00352.x

Press, 2010, Explaining variability in ciclosporin exposure in adult kidney transplant recipients, Eur J Clin Pharmacol., 66, 579, 10.1007/s00228-010-0810-9

Cremers, 2003, A compartmental pharmacokinetic model of cyclosporin and its predictive performance after Bayesian estimation in kidney and simultaneous pancreas-kidney transplant recipients, Nephrol Dial Transpl., 18, 1201, 10.1093/ndt/gfg065

Johnston, 2000, Pharmacokinetic validation of neoral absorption profiling, Transpl Proc., 32, 53S, 10.1016/S0041-1345(00)00864-2

Vester, 2004, Absorption phase cyclosporine (C(2h)) monitoring in the first weeks after pediatric renal transplantation, Pediatr Nephrol., 19, 1273, 10.1007/s00467-004-1617-7

Napoli, 1996, Routine clinical monitoring of sirolimus (rapamycin) whole-blood concentrations by HPLC with ultraviolet detection, Clin Chem., 42, 1943, 10.1093/clinchem/42.12.1943

van Gelder, 2001, Comparison of the effects of tacrolimus and cyclosporine on the pharmacokinetics of mycophenolic acid, Ther Drug Monit., 23, 119, 10.1097/00007691-200104000-00005

Mulley, 2012, Mycophenolate and lower graft function reduce the seroresponse of kidney transplant recipients to pandemic H1N1 vaccination, Kidney Int., 82, 212, 10.1038/ki.2012.106

Resende, 2013, Low seroconversion after one dose of AS03-adjuvanted H1N1 pandemic influenza vaccine in solid-organ transplant recipients, Can J Infect Dis Med Microbiol., 24, e7, 10.1155/2013/256756

Tett, 2011, Mycophenolate, clinical pharmacokinetics, formulations, and methods for assessing drug exposure, Transpl Rev (Orlando)., 25, 47, 10.1016/j.trre.2010.06.001

Wood, 2015, Adrenal insufficiency as a result of ritonavir and exogenous steroid exposure: report of 6 cases and recommendation for management, J Int Assoc Provid AIDS Care., 14, 300, 10.1177/2325957414567681

Busse, 2008, Influence of antiretroviral drugs on the pharmacokinetics of prednisolone in HIV-infected individuals, J Acquir Immune Defic Syndr., 48, 561, 10.1097/QAI.0b013e31817bebeb

Zumla, 2015, Middle East respiratory syndrome, Lancet., 386, 995, 10.1016/S0140-6736(15)60454-8

Arabi, 2018, Corticosteroid therapy for critically ill patients with Middle East respiratory syndrome, Am J Respir Crit Care Med., 197, 757, 10.1164/rccm.201706-1172OC

Russell, 2020, Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury, Lancet., 395, 473, 10.1016/S0140-6736(20)30317-2

Shang, 2020, On the use of corticosteroids for 2019-nCoV pneumonia, Lancet., 395, 683, 10.1016/S0140-6736(20)30361-5

Warren, 2016, Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys, Nature., 531, 381, 10.1038/nature17180

Wang, 2020, Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro, Cell Res, 30, 269, 10.1038/s41422-020-0282-0

Huang, 2020, Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China, Lancet., 395, 497, 10.1016/S0140-6736(20)30183-5

Kaneko, 2018, Tocilizumab discontinuation after attaining remission in patients with rheumatoid arthritis who were treated with tocilizumab alone or in combination with methotrexate: results from a prospective randomised controlled study (the second year of the SURPRISE study), Ann Rheum Dis., 77, 1268, 10.1136/annrheumdis-2018-213416

Stone, 2017, Trial of tocilizumab in giant-cell arteritis, N Engl J Med., 377, 317, 10.1056/NEJMoa1613849

Long, 2016, Modeling therapeutic antibody-small molecule drug-drug interactions using a three-dimensional perfusable human liver coculture platform, Drug Metab Dispos., 44, 1940, 10.1124/dmd.116.071456

Sheppard, 2017, Tocilizumab (actemra), Hum Vaccin Immunother., 13, 1972, 10.1080/21645515.2017.1316909

Tu, 2014, An interdisciplinary approach for renal transplant recipients with severe pneumonia: a single ICU experience, Intensive Care Med., 40, 914, 10.1007/s00134-014-3296-6

Zhu, 2020, Successful recovery of COVID-19 pneumonia in a renal transplant recipient with long-term immunosuppression, Am J Transpl., 10.1111/ajt.15869

Guillen, 2020, Case report of COVID-19 in a kidney transplant recipient: does immunosuppression alter the clinical presentation?, Am J Transpl., 10.1111/ajt.15874

Ventura-Aguiar, 2016, Safety of mTOR inhibitors in adult solid organ transplantation, Expert Opin Drug Saf., 15, 303, 10.1517/14740338.2016.1132698

Tielemans, 2019, Immunosuppressive drugs and the gastrointestinal tract in renal transplant patients, Transpl Rev (Orlando)., 33, 55, 10.1016/j.trre.2018.11.001

Harvey, 2014, Cancer, inflammation, and therapy: effects on cytochrome p450-mediated drug metabolism and implications for novel immunotherapeutic agents, Clin Pharmacol Ther., 96, 449, 10.1038/clpt.2014.143