1
|
Kale A, Shelke V, Dagar N, Anders HJ, Gaikwad AB. How to use COVID-19 antiviral drugs in patients with chronic kidney disease. Front Pharmacol 2023; 14:1053814. [PMID: 36843922 PMCID: PMC9947246 DOI: 10.3389/fphar.2023.1053814] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Antiviral drugs such as Remdesivir (Veklury), Nirmatrelvir with Ritonavir (Paxlovid), Azvudine, and Molnupiravir (Lagevrio) can reduce the risk for severe and fatal Coronavirus Disease (COVID)-19. Although chronic kidney disease is a highly prevalent risk factor for severe and fatal COVID-19, most clinical trials with these drugs excluded patients with impaired kidney function. Advanced CKD is associated with a state of secondary immunodeficiency (SIDKD), which increases the susceptibility to severe COVID-19, COVID-19 complications, and the risk of hospitalization and mortality among COVID-19 patients. The risk to develop COVID-19 related acute kidney injury is higher in patients with precedent CKD. Selecting appropriate therapies for COVID-19 patients with impaired kidney function is a challenge for healthcare professionals. Here, we discuss the pharmacokinetics and pharmacodynamics of COVID-19-related antiviral drugs with a focus on their potential use and dosing in COVID-19 patients with different stages of CKD. Additionally, we describe the adverse effects and precautions to be taken into account when using these antivirals in COVID-19 patients with CKD. Lastly, we also discuss about the use of monoclonal antibodies in COVID-19 patients with kidney disease and related complications.
Collapse
Affiliation(s)
- Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Vishwadeep Shelke
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Neha Dagar
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| |
Collapse
|
2
|
Sharma A, Sharma P, Kapila I, Abbot V. A Combination of Novel HIV-1 Protease Inhibitor and Cytochrome P450 (CYP) Enzyme Inhibitor to Explore the Future Prospective of Antiviral Agents: Evotaz. Curr HIV Res 2023; 21:149-159. [PMID: 37221692 DOI: 10.2174/1570162x21666230522123631] [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: 11/26/2022] [Revised: 03/27/2023] [Accepted: 04/19/2023] [Indexed: 05/25/2023]
Abstract
Viruses belong to the class of micro-organisms that are well known for causing infections in the human body. Antiviral medications are given out to prevent the spread of disease-causing viruses. When the viruses are actively reproducing, these agents have their greatest impact. It is particularly challenging to develop virus-specific medications since viruses share the majority of the metabolic functions of the host cell. In the continuous search for better antiviral agents, the United States Food and Drug Administration (USFDA) approved a new drug named Evotaz on January 29, 2015 for the treatment of human immunodeficiency virus (HIV). Evotaz is a combined once-daily fixed drug, containing Atazanavir, an HIV protease inhibitor, and cobicistat, an inhibitor of the human liver cytochrome P450 (CYP) enzyme. The medication is created such that it can kill viruses by concurrently inhibiting protease and CYP enzymes. The medicine is still being studied for a number of criteria, but its usefulness in children under the age of 12 is currently unknown. The preclinical and clinical characteristics of Evotaz, as well as its safety and efficacy profiles and a comparison of the novel drug with antiviral medications presently available in the market, are the main topics of this review paper.
Collapse
Affiliation(s)
- Abha Sharma
- Faculty of Pharmaceutical Sciences, PCTE Group of Institutes, Campus-2, Near Baddowal Cantt., Ferozpur Road, Ludhiana, 142021, India
| | - Poonam Sharma
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, 173234, Himachal Pradesh, India
| | - Isha Kapila
- Pharmaceutical Chemistry Department, Chandigarh College of Pharmacy, Landran, Mohali, 140307, Punjab, India
| | - Vikrant Abbot
- Department of Pharmaceutical Sciences, Saraswati Group of Colleges, Gharuan, Mohali, 140413, Punjab, India
| |
Collapse
|
3
|
Viral proteases as therapeutic targets. Mol Aspects Med 2022; 88:101159. [PMID: 36459838 PMCID: PMC9706241 DOI: 10.1016/j.mam.2022.101159] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022]
Abstract
Some medically important viruses-including retroviruses, flaviviruses, coronaviruses, and herpesviruses-code for a protease, which is indispensable for viral maturation and pathogenesis. Viral protease inhibitors have become an important class of antiviral drugs. Development of the first-in-class viral protease inhibitor saquinavir, which targets HIV protease, started a new era in the treatment of chronic viral diseases. Combining several drugs that target different steps of the viral life cycle enables use of lower doses of individual drugs (and thereby reduction of potential side effects, which frequently occur during long term therapy) and reduces drug-resistance development. Currently, several HIV and HCV protease inhibitors are routinely used in clinical practice. In addition, a drug including an inhibitor of SARS-CoV-2 main protease, nirmatrelvir (co-administered with a pharmacokinetic booster ritonavir as Paxlovid®), was recently authorized for emergency use. This review summarizes the basic features of the proteases of human immunodeficiency virus (HIV), hepatitis C virus (HCV), and SARS-CoV-2 and discusses the properties of their inhibitors in clinical use, as well as development of compounds in the pipeline.
Collapse
|
4
|
Al-Taie A, Büyük AŞ, Sardas S. Considerations into pharmacogenomics of COVID-19 pharmacotherapy: Hope, hype and reality. Pulm Pharmacol Ther 2022; 77:102172. [PMID: 36265833 PMCID: PMC9576910 DOI: 10.1016/j.pupt.2022.102172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/16/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
Abstract
COVID-19 medicines, such as molnupiravir are beginning to emerge for public health and clinical practice. On the other hand, drugs display marked variability in their efficacy and safety. Hence, COVID-19 medicines, as with all drugs, will be subject to the age-old maxim "one size prescription does not fit all". In this context, pharmacogenomics is the study of genome-by-drug interactions and offers insights on mechanisms of patient-to-patient and between-population variations in drug efficacy and safety. Pharmacogenomics information is crucial to tailoring the patients' prescriptions to achieve COVID-19 preventive and therapeutic interventions that take into account the host biology, patients' genome, and variable environmental exposures that collectively influence drug efficacy and safety. This expert review critically evaluates and summarizes the pharmacogenomics and personalized medicine aspects of the emerging COVID-19 drugs, and other selected drug interventions deployed to date. Here, we aim to sort out the hope, hype, and reality and suggest that there are veritable prospects to advance COVID-19 medicines for public health benefits, provided that pharmacogenomics is considered and implemented adequately. Pharmacogenomics is an integral part of rational and evidence-based medical practice. Scientists, health care professionals, pharmacists, pharmacovigilance practitioners, and importantly, patients stand to benefit by expanding the current pandemic response toolbox by the science of pharmacogenomics, and its applications in COVID-19 medicines and clinical trials.
Collapse
Affiliation(s)
- Anmar Al-Taie
- Clinical Pharmacy Department, Faculty of Pharmacy, Istinye University, Istanbul, Turkey.
| | - Ayşe Şeyma Büyük
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istinye University, Istanbul, Turkey
| | - Semra Sardas
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istinye University, Istanbul, Turkey
| |
Collapse
|
5
|
Franczyk B, Rysz J, Miłoński J, Konecki T, Rysz-Górzyńska M, Gluba-Brzózka A. Will the Use of Pharmacogenetics Improve Treatment Efficiency in COVID-19? Pharmaceuticals (Basel) 2022; 15:739. [PMID: 35745658 PMCID: PMC9230944 DOI: 10.3390/ph15060739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
The COVID-19 pandemic is associated with a global health crisis and the greatest challenge for scientists and doctors. The virus causes severe acute respiratory syndrome with an outcome that is fatal in more vulnerable populations. Due to the need to find an efficient treatment in a short time, there were several drugs that were repurposed or repositioned for COVID-19. There are many types of available COVID-19 therapies, including antiviral agents (remdesivir, lopinavir/ritonavir, oseltamivir), antibiotics (azithromycin), antiparasitics (chloroquine, hydroxychloroquine, ivermectin), and corticosteroids (dexamethasone). A combination of antivirals with various mechanisms of action may be more efficient. However, the use of some of these medicines can be related to the occurrence of adverse effects. Some promising drug candidates have been found to be ineffective in clinical trials. The knowledge of pharmacogenetic issues, which translate into variability in drug conversion from prodrug into drug, metabolism as well as transport, could help to predict treatment efficiency and the occurrence of adverse effects in patients. However, many drugs used for the treatment of COVID-19 have not undergone pharmacogenetic studies, perhaps as a result of the lack of time.
Collapse
Affiliation(s)
- Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| | - Jarosław Miłoński
- Department of Otolaryngology, Laryngological Oncology, Audiology and Phoniatrics, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Tomasz Konecki
- Department of Urology, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Magdalena Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (B.F.); (J.R.)
| |
Collapse
|
6
|
Li X, Peng T. Strategy, Progress, and Challenges of Drug Repurposing for Efficient Antiviral Discovery. Front Pharmacol 2021; 12:660710. [PMID: 34017257 PMCID: PMC8129523 DOI: 10.3389/fphar.2021.660710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
Emerging or re-emerging viruses are still major threats to public health. Prophylactic vaccines represent the most effective way to prevent virus infection; however, antivirals are more promising for those viruses against which vaccines are not effective enough or contemporarily unavailable. Because of the slow pace of novel antiviral discovery, the high disuse rates, and the substantial cost, repurposing of the well-characterized therapeutics, either approved or under investigation, is becoming an attractive strategy to identify the new directions to treat virus infections. In this review, we described recent progress in identifying broad-spectrum antivirals through drug repurposing. We defined the two major categories of the repurposed antivirals, direct-acting repurposed antivirals (DARA) and host-targeting repurposed antivirals (HTRA). Under each category, we summarized repurposed antivirals with potential broad-spectrum activity against a variety of viruses and discussed the possible mechanisms of action. Finally, we proposed the potential investigative directions of drug repurposing.
Collapse
Affiliation(s)
- Xinlei Li
- State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, College of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Tao Peng
- State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, College of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Pharmacogenomics and COVID-19: clinical implications of human genome interactions with repurposed drugs. THE PHARMACOGENOMICS JOURNAL 2021; 21:275-284. [PMID: 33542445 PMCID: PMC7859465 DOI: 10.1038/s41397-021-00209-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 12/07/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
Abstract
The outbreak of Coronavirus disease 2019 (COVID-19) has evolved into an emergent global pandemic. Many drugs without established efficacy are being used to treat COVID-19 patients either as an offlabel/compassionate use or as a clinical trial. Although drug repurposing is an attractive approach with reduced time and cost, there is a need to make predictions on success before the start of therapy. For the optimum use of these repurposed drugs, many factors should be considered such as drug–gene or dug–drug interactions, drug toxicity, and patient co-morbidity. There is limited data on the pharmacogenomics of these agents and this may constitute an obstacle for successful COVID-19 therapy. This article reviewed the available human genome interactions with some promising repurposed drugs for COVID-19 management. These drugs include chloroquine (CQ), hydroxychloroquine (HCQ), azithromycin, lopinavir/ritonavir (LPV/r), atazanavir (ATV), favipiravir (FVP), nevirapine (NVP), efavirenz (EFV), oseltamivir, remdesivir, anakinra, tocilizumab (TCZ), eculizumab, heme oxygenase 1 (HO-1) regulators, renin–angiotensin–aldosterone system (RAAS) inhibitors, ivermectin, and nitazoxanide. Drug-gene variant pairs that may alter the therapeutic outcomes in COVID-19 patients are presented. The major drug variant pairs that associated with variations in clinical efficacy include CQ/HCQ (CYP2C8, CYP2D6, ACE2, and HO-1); azithromycin (ABCB1); LPV/r (SLCO1B1, ABCB1, ABCC2 and CYP3A); NVP (ABCC10); oseltamivir (CES1 and ABCB1); remdesivir (CYP2C8, CYP2D6, CYP3A4, and OATP1B1); anakinra (IL-1a); and TCZ (IL6R and FCGR3A). The major drug variant pairs that associated with variations in adverse effects include CQ/HCQ (G6PD; hemolysis and ABCA4; retinopathy), ATV (MDR1 and UGT1A1*28; hyperbilirubinemia; and APOA5; dyslipidemia), NVP (HLA-DRB1*01, HLA-B*3505 and CYP2B6; skin rash and MDR1; hepatotoxicity), and EFV (CYP2B6; depression and suicidal tendencies).
Collapse
|
8
|
Robles NR, Fici F, Valladares J, Grassi G. Antiretroviral Treatment and Antihypertensive Therapy. Curr Pharm Des 2021; 27:4116-4124. [PMID: 34784859 DOI: 10.2174/1381612827666210810090805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 05/10/2021] [Indexed: 11/22/2022]
Abstract
The presence of hypertension among the population with human immunodeficiency virus (HIV) has become a new threat to the health and well-being of people living with this disease, in particular, among those who received antiretroviral therapy. The estimated prevalence of high blood pressure in HIV-infected patients is significantly higher than the rate observed in HIV-uninfected subjects. The approach to the HIV-positive patient requires the assessment of individual cardiovascular risk and its consideration when designing the individualized target. On the other hand, the numerous pharmacological interactions of antiretroviral (ARV) drugs are essential elements to take into account. Serum levels of any kind of antihypertensive drugs may be influenced by the coadministration of protease inhibitors, non-nucleoside reverse transcriptase inhibitor, or other antiretroviral. Similarly, plasma concentrations of antiretroviral drugs can be increased by the concomitant use of calcium channel blockers or diuretics. In this regard, the treatment of high blood pressure in HIV patients should be preferentially based on ACE inhibitors or thiazide/thiazide-like diuretics or their combination.
Collapse
Affiliation(s)
- Nicolás R Robles
- Servicio de Nefrologia, Hospital Universitario de Badajoz, Badajoz, Spain
| | - Francesco Fici
- Cardiovascular Risk Chair, University of Salamanca School of Medicine, Salamanca, Spain
| | - Julian Valladares
- Servicio de Nefrologia, Hospital Universitario de Badajoz, Badajoz, Spain
| | - Guido Grassi
- Clinica Medica, Universita Milano-Bicocca, Milan, Spain
| |
Collapse
|
9
|
de Almeida SMV, Santos Soares JC, Dos Santos KL, Alves JEF, Ribeiro AG, Jacob ÍTT, da Silva Ferreira CJ, Dos Santos JC, de Oliveira JF, de Carvalho Junior LB, de Lima MDCA. COVID-19 therapy: What weapons do we bring into battle? Bioorg Med Chem 2020; 28:115757. [PMID: 32992245 PMCID: PMC7481143 DOI: 10.1016/j.bmc.2020.115757] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/29/2020] [Accepted: 09/03/2020] [Indexed: 01/18/2023]
Abstract
Urgent treatments, in any modality, to fight SARS-CoV-2 infections are desired by society in general, by health professionals, by Estate-leaders and, mainly, by the scientific community, because one thing is certain amidst the numerous uncertainties regarding COVID-19: knowledge is the means to discover or to produce an effective treatment against this global disease. Scientists from several areas in the world are still committed to this mission, as shown by the accelerated scientific production in the first half of 2020 with over 25,000 published articles related to the new coronavirus. Three great lines of publications related to COVID-19 were identified for building this article: The first refers to knowledge production concerning the virus and pathophysiology of COVID-19; the second regards efforts to produce vaccines against SARS-CoV-2 at a speed without precedent in the history of science; the third comprehends the attempts to find a marketed drug that can be used to treat COVID-19 by drug repurposing. In this review, the drugs that have been repurposed so far are grouped according to their chemical class. Their structures will be presented to provide better understanding of their structural similarities and possible correlations with mechanisms of actions. This can help identifying anti-SARS-CoV-2 promising therapeutic agents.
Collapse
Affiliation(s)
- Sinara Mônica Vitalino de Almeida
- Laboratório de Biologia Molecular, Universidade de Pernambuco, Garanhuns, PE, Brazil; Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil; Laboratório de Imunopatologia Keizo Asami (LIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil.
| | - José Cleberson Santos Soares
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Keriolaine Lima Dos Santos
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Amélia Galdino Ribeiro
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Íris Trindade Tenório Jacob
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | | | - Jamerson Ferreira de Oliveira
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Maria do Carmo Alves de Lima
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| |
Collapse
|
10
|
Ong CWM, Migliori GB, Raviglione M, MacGregor-Skinner G, Sotgiu G, Alffenaar JW, Tiberi S, Adlhoch C, Alonzi T, Archuleta S, Brusin S, Cambau E, Capobianchi MR, Castilletti C, Centis R, Cirillo DM, D'Ambrosio L, Delogu G, Esposito SMR, Figueroa J, Friedland JS, Ho BCH, Ippolito G, Jankovic M, Kim HY, Rosales Klintz S, Ködmön C, Lalle E, Leo YS, Leung CC, Märtson AG, Melazzini MG, Najafi Fard S, Penttinen P, Petrone L, Petruccioli E, Pontali E, Saderi L, Santin M, Spanevello A, van Crevel R, van der Werf MJ, Visca D, Viveiros M, Zellweger JP, Zumla A, Goletti D. Epidemic and pandemic viral infections: impact on tuberculosis and the lung: A consensus by the World Association for Infectious Diseases and Immunological Disorders (WAidid), Global Tuberculosis Network (GTN), and members of the European Society of Clinical Microbiology and Infectious Diseases Study Group for Mycobacterial Infections (ESGMYC). Eur Respir J 2020; 56:2001727. [PMID: 32586885 PMCID: PMC7527651 DOI: 10.1183/13993003.01727-2020] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 01/08/2023]
Abstract
Major epidemics, including some that qualify as pandemics, such as severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), HIV, influenza A (H1N1)pdm/09 and most recently COVID-19, affect the lung. Tuberculosis (TB) remains the top infectious disease killer, but apart from syndemic TB/HIV little is known regarding the interaction of viral epidemics and pandemics with TB. The aim of this consensus-based document is to describe the effects of viral infections resulting in epidemics and pandemics that affect the lung (MERS, SARS, HIV, influenza A (H1N1)pdm/09 and COVID-19) and their interactions with TB. A search of the scientific literature was performed. A writing committee of international experts including the European Centre for Disease Prevention and Control Public Health Emergency (ECDC PHE) team, the World Association for Infectious Diseases and Immunological Disorders (WAidid), the Global Tuberculosis Network (GTN), and members of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Mycobacterial Infections (ESGMYC) was established. Consensus was achieved after multiple rounds of revisions between the writing committee and a larger expert group. A Delphi process involving the core group of authors (excluding the ECDC PHE team) identified the areas requiring review/consensus, followed by a second round to refine the definitive consensus elements. The epidemiology and immunology of these viral infections and their interactions with TB are discussed with implications for diagnosis, treatment and prevention of airborne infections (infection control, viral containment and workplace safety). This consensus document represents a rapid and comprehensive summary on what is known on the topic.
Collapse
Affiliation(s)
- Catherine Wei Min Ong
- Dept of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
- These authors contributed equally
- Members of ESGMYC
| | - Giovanni Battista Migliori
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
- These authors contributed equally
| | - Mario Raviglione
- Centre for Multidisciplinary Research in Health Science, University of Milan, Milan, Italy
- Global Studies Institute, University of Geneva, Geneva, Switzerland
| | | | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Dept of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Jan-Willem Alffenaar
- Sydney Pharmacy School, University of Sydney, Sydney, Australia
- Westmead Hospital, Sydney, Australia
- Marie Bashir Institute of Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
- Members of ESGMYC
| | - Simon Tiberi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Division of Infection, Royal London Hospital, Barts Health NHS Trust, London, UK
- Members of ESGMYC
| | - Cornelia Adlhoch
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Tonino Alonzi
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Sophia Archuleta
- Dept of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sergio Brusin
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Emmanuelle Cambau
- AP-HP-Lariboisiere, Bacteriologie, Laboratory Associated to the National Reference Centre for Mycobacteria, IAME UMR1137, INSERM, University of Paris, Paris, France
- Members of ESGMYC
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Rosella Centis
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Members of ESGMYC
| | | | - Giovanni Delogu
- Università Cattolica Sacro Cuore, Roma, Italy
- Mater Olbia Hospital, Olbia, Italy
- Members of ESGMYC
| | - Susanna M R Esposito
- Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | | | - Jon S Friedland
- St George's, University of London, London, UK
- Members of ESGMYC
| | - Benjamin Choon Heng Ho
- Tuberculosis Control Unit, Dept of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore
| | - Giuseppe Ippolito
- Scientific Direction, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Mateja Jankovic
- School of Medicine, University of Zagreb and Clinic for Respiratory Diseases, University Hospital Center Zagreb, Zagreb, Croatia
- Members of ESGMYC
| | - Hannah Yejin Kim
- Sydney Pharmacy School, University of Sydney, Sydney, Australia
- Westmead Hospital, Sydney, Australia
- Marie Bashir Institute of Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia
| | - Senia Rosales Klintz
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Csaba Ködmön
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Eleonora Lalle
- Laboratory of Virology, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Yee Sin Leo
- National Centre for Infectious Diseases, Singapore
| | - Chi-Chiu Leung
- Hong Kong Tuberculosis, Chest and Heart Diseases Association, Wanchai, Hong Kong, China
| | - Anne-Grete Märtson
- Dept of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Saeid Najafi Fard
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Pasi Penttinen
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Linda Petrone
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | - Elisa Petruccioli
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
| | | | - Laura Saderi
- Clinical Epidemiology and Medical Statistics Unit, Dept of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Miguel Santin
- Dept of Infectious Diseases, Bellvitge University Hospital-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Dept of Clinical Science, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Members of ESGMYC
| | - Antonio Spanevello
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy
- Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Reinout van Crevel
- Radboudumc Center for Infectious Diseases, Radboud Institute for Health Sciences, Radboudumc, Nijmegen, The Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Dept of Medicine, University of Oxford, Oxford, UK
- Members of ESGMYC
| | - Marieke J van der Werf
- Public Health Emergency Team, European Centre for Disease Prevention and Control, Stockholm, Sweden
- European Centre for Disease Prevention and Control Public Health Emergency team co-authors
| | - Dina Visca
- Division of Pulmonary Rehabilitation, Istituti Clinici Scientifici Maugeri, IRCCS, Tradate, Italy
- Dept of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Miguel Viveiros
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, NOVA University of Lisbon, Lisbon, Portugal
- Members of ESGMYC
| | | | - Alimuddin Zumla
- Dept of Infection, Division of Infection and Immunity, University College London and NIHR Biomedical Research Centre, UCL Hospitals NHS Foundation Trust, London, UK
| | - Delia Goletti
- Translational Research Unit, Epidemiology and Preclinical Research Dept, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS, Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, Rome, Italy
- Members of ESGMYC
| |
Collapse
|
11
|
Joe MB, Roland L, Laurent C, Patrick LM, Sawoo O, Gaston TL, Bruno E, Gilles P, Philippe P. Concomitant Use of Cotrimoxazole and Atazanavir in HIV-infected Patients: A Therapeutic Drug Monitoring and Pharmacovigilance Based Dual Approach. ACTA ACUST UNITED AC 2020; 14:214-223. [PMID: 30961507 DOI: 10.2174/1574884714666190405160612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Cotrimoxazole is the main antibiotic used in primary prophylaxis for opportunistic infections in advanced HIV infection. This drug can inhibit one of the metabolic pathways of atazanavir (ATV), such as the cytochromes P450 (CYP) 2C8/2C9 and could interfere with its safety and efficacy. OBJECTIVE We studied the drug-drug interaction (DDI) between cotrimoxazole and ATV by using therapeutic drug monitoring (TDM) and pharmacovigilance (PV) approaches. METHODS We compared a group of patients treated with cotrimoxazole and receiving an ATV-based regimen to controls. This historical cohort analysis used data from Dat'AIDS in HIV-infected patients who had at least two lowest plasma concentrations (C-trough) of ATV during their outpatient follow-up. Likewise, we used the international pharmacovigilance data from VigiBase to evaluate the notifications of hyperbilirubinemia reported with ATV. RESULTS In the TDM analysis, the two groups of patients (treated with cotrimoxazole and controls) were almost homogeneous concerning the main baseline features. After at least six months of ATVbased regimen, there was no significant difference in the safety threshold of the ATV C-trough [with an adjusted odds ratio (aOR) of 1.4 (95% CI: 0.5 - 4.4)] compared to controls. We observed similar results with the efficacy thresholds of ATV C-trough. Regarding the PV analysis, there was no difference in hyperbilirubinemia occurring with ATV when cotrimoxazole was concomitant, with an adjusted reporting odds ratio (aROR) of 0.9 (95% CI: 0.6 to 1.2). CONCLUSION This study showed a relevant concomitant use between Cotrimoxazole and ATV based on TDM and PV approaches.
Collapse
Affiliation(s)
- Miantezila B Joe
- EA4065 Ecosystème Intestinal, Probiotique, Antibiotiques, Université Paris Descartes SPC, Paris, France.,Unité de Pharmacologie Clinique, Université de Kinshasa, Kinshasa, RD, Congo.,Centre Régional de Pharmacovigilance, Hôpital Cochin, Assistance publique- Hopitaux de Paris, Paris, France
| | - Landman Roland
- Service des Maladies Infectieuses et Tropicales, Hôpital Bichat, Paris, France.,IAME, UMR 1137, Université Paris Diderot, Sorbonne Paris Cité and INSERM, Paris, France
| | - Chouchana Laurent
- Centre Régional de Pharmacovigilance, Hôpital Cochin, Assistance publique- Hopitaux de Paris, Paris, France.,EA7323 Evaluation thérapeutique et pharmacologie périnatale et pédiatrique, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Lê M Patrick
- IAME, UMR 1137, Université Paris Diderot, Sorbonne Paris Cité and INSERM, Paris, France.,AP-HP, Hôpital Bichat-Claude Bernard, Laboratoire de Pharmaco-Toxicologie, Paris, France
| | - Olivier Sawoo
- EA4065 Ecosystème Intestinal, Probiotique, Antibiotiques, Université Paris Descartes SPC, Paris, France.,Microbial Ecology Laboratory (MIEL), CNAM, Paris, France
| | - Tona L Gaston
- Unité de Pharmacologie Clinique, Université de Kinshasa, Kinshasa, RD, Congo
| | - Eto Bruno
- TBC TransCell-Lab, Faculté de Médecine Xavier Bichat, Paris, France
| | - Peytavin Gilles
- IAME, UMR 1137, Université Paris Diderot, Sorbonne Paris Cité and INSERM, Paris, France.,AP-HP, Hôpital Bichat-Claude Bernard, Laboratoire de Pharmaco-Toxicologie, Paris, France
| | - Pochart Philippe
- EA4065 Ecosystème Intestinal, Probiotique, Antibiotiques, Université Paris Descartes SPC, Paris, France.,Microbial Ecology Laboratory (MIEL), CNAM, Paris, France
| |
Collapse
|
12
|
Cirrincione LR, Senneker T, Scarsi K, Tseng A. Drug Interactions with Gender-Affirming Hormone Therapy: Focus on Antiretrovirals and Direct Acting Antivirals. Expert Opin Drug Metab Toxicol 2020; 16:565-582. [PMID: 32479127 DOI: 10.1080/17425255.2020.1777278] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Gender-affirming care may include hormonal therapy to attain desired health outcomes in transgender (trans) individuals. To provide safe, affirming medical care for trans patients, health care providers must identify and manage drug-drug interactions (DDIs) between gender affirming hormonal therapy (GAHT) and other medication therapies. AREAS COVERED This review summarizes available data on DDIs between GAHT and antiretrovirals (ARVs) or hepatitis C direct acting antivirals (DAAs). Potential pharmacokinetic and pharmacodynamic DDIs are predicted based on GAHT, ARV, and DAA pharmacology and adverse event profiles. Clinical management strategies are discussed. EXPERT OPINION GAHT may be involved in pharmacokinetic and/or pharmacodynamic DDIs. Certain ARV classes (non-nucleoside reverse transcriptase inhibitors, protease inhibitors) may alter GAHT disposition, whereas selected ARVs (unboosted integrase inhibitors, doravirine, or rilpivirine) may have less impact on GAHT. DAAs may interact with GAHT, but the clinical relevance is unclear. ARV- and/or DAA-associated side effects (including depression, cardiovascular disease, hyperlipidemia) are important to consider in the clinical management of trans patients. Clinicians must evaluate potential DDIs and overlapping side effects between ARVs, DAAs and GAHT when providing care for trans patients.
Collapse
Affiliation(s)
- Lauren R Cirrincione
- Department of Pharmacy, University of Washington School of Pharmacy , Seattle, WA, USA
| | - Tessa Senneker
- Department of Pharmacy, Kingston General Hospital , Kingston, ON, Canada
| | - Kimberly Scarsi
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center , Omaha, NE, USA
| | - Alice Tseng
- Faculty of Pharmacy, University of Toronto , Toronto, ON, Canada.,University Health Network , Toronto, ON, Canada
| |
Collapse
|
13
|
Singh H, Lata S, Choudhari R, Dhole TN. Prevalence of ABCC3-1767G/A polymorphism among patients with antiretroviral-associated hepatotoxicity. Mol Genet Genomic Med 2020; 8:e1124. [PMID: 32212330 PMCID: PMC7284032 DOI: 10.1002/mgg3.1124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022] Open
Abstract
Background Plasma concentrations of antiretrovirals (ARVs) regimens have considerably varied in individuals of human immunodeficiency virus (HIV) because of variations in the expression of drug‐metabolizing and transporter genes. Transporter genes play an important role in the disposition of drugs. Polymorphism in transporter gene (ABCC3) affects the MRP3 expression and varies the treatment outcome. Method We examined the polymorphism of ABCC3‐1767G/A gene in a total of 165 HIV patients (out of 165 HIV patients, 34 were with and 131 were without hepatotoxicity) and 156 healthy individuals using the polymerase chain reaction–restriction fragment length polymorphism method. Results In univariate analysis, we found a decreased prevalence of ABCC3 1767GA, 1767GA+AA genotypes, and 1767A allele in patients with hepatotoxicity as compared to patients without hepatotoxicity (23.5% vs. 28.2% and 23.5% vs. 30.53%; 11.76% vs. 16.41%), while a higher prevalence of 1767AA genotype was observed in HIV patients in comparison with healthy controls (2.3% vs. 1.3%, odds ratio [OR] = 1.71, 95% confidence interval [CI]: 0.23–15.03, p = .89). The frequency of ABCC3‐1767AA genotype was dispersed higher in individuals with early and advanced HIV disease stage in comparison with healthy controls (5.3% vs. 1.3%, OR = 4.73, p = .70; 8.9% vs. 1.3%, OR = 1.89, p = .91). A higher occurrence of ABCC3‐1767AA genotype was found in tobacco using HIV patients without hepatotoxicity compared with nonusers (4.7% vs. 1.1%, OR = 4.28, p = .52). The distribution of ABCC3‐1767GA genotype was higher in nevirapine receiving HIV patients irrespective of their hepatotoxicity status as compared to nonusers (30.4% vs. 9.1%, OR = 3.34, p = .22; 29.4% vs. 16.7%, OR = 1.69, p = .77). In multivariate analysis, HIV patients receiving nevirapine and with hepatotoxicity was found to have a significant risk for severity of hepatotoxicity (OR = 4.56, 95% CI: 1.60–12.99, p = .004). Conclusion ABCC3 1767G/A polymorphism was not significantly associated with susceptibility to ARV‐associated hepatotoxicity, although ABCC3 1767AA genotype designated a risk for acquisition of hepatotoxicity and advancement of the disease. Nevirapine usage emerged as an independent risk factor for hepatotoxicity severity.
Collapse
Affiliation(s)
- HariOm Singh
- Department of Molecular Biology, National AIDS Research Institute, Pune, India
| | - Sonam Lata
- Department of Molecular Biology, National AIDS Research Institute, Pune, India
| | - Ranjana Choudhari
- Department of Clinical Epidemiology, National Institute of Occupational Health, Ahmedabad, India
| | - Tapan N Dhole
- Department of Microbiology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
14
|
de Man FM, Goey AKL, van Schaik RHN, Mathijssen RHJ, Bins S. Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics. Clin Pharmacokinet 2019. [PMID: 29520731 PMCID: PMC6132501 DOI: 10.1007/s40262-018-0644-7] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Since its clinical introduction in 1998, the topoisomerase I inhibitor irinotecan has been widely used in the treatment of solid tumors, including colorectal, pancreatic, and lung cancer. Irinotecan therapy is characterized by several dose-limiting toxicities and large interindividual pharmacokinetic variability. Irinotecan has a highly complex metabolism, including hydrolyzation by carboxylesterases to its active metabolite SN-38, which is 100- to 1000-fold more active compared with irinotecan itself. Several phase I and II enzymes, including cytochrome P450 (CYP) 3A4 and uridine diphosphate glucuronosyltransferase (UGT) 1A, are involved in the formation of inactive metabolites, making its metabolism prone to environmental and genetic influences. Genetic variants in the DNA of these enzymes and transporters could predict a part of the drug-related toxicity and efficacy of treatment, which has been shown in retrospective and prospective trials and meta-analyses. Patient characteristics, lifestyle and comedication also influence irinotecan pharmacokinetics. Other factors, including dietary restriction, are currently being studied. Meanwhile, a more tailored approach to prevent excessive toxicity and optimize efficacy is warranted. This review provides an updated overview on today’s literature on irinotecan pharmacokinetics, pharmacodynamics, and pharmacogenetics.
Collapse
Affiliation(s)
- Femke M de Man
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands
| | - Andrew K L Goey
- Department of Hospital Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Wick S, Walsh DI, Bobrow J, Hamad-Schifferli K, Kong DS, Thorsen T, Mroszczyk K, Carr PA. PERSIA for Direct Fluorescence Measurements of Transcription, Translation, and Enzyme Activity in Cell-Free Systems. ACS Synth Biol 2019; 8:1010-1025. [PMID: 30920800 PMCID: PMC6830305 DOI: 10.1021/acssynbio.8b00450] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Quantification of biology's central dogma (transcription and translation) is pursued by a variety of methods. Direct, immediate, and ongoing quantification of these events is difficult to achieve. Common practice is to use fluorescent or luminescent proteins to report indirectly on prior cellular events, such as turning on a gene in a genetic circuit. We present an alternative approach, PURExpress-ReAsH-Spinach In-vitro Analysis (PERSIA). PERSIA provides information on the production of RNA and protein during cell-free reactions by employing short RNA and peptide tags. Upon synthesis, these tags yield quantifiable fluorescent signal without interfering with other biochemical events. We demonstrate the applicability of PERSIA in measuring cell-free transcription, translation, and other enzymatic activity in a variety of applications: from sequence-structure-function studies, to genetic code engineering, to testing antiviral drug resistance.
Collapse
Affiliation(s)
- Scott Wick
- MIT Lincoln Laboratory , Lexington , Massachusetts 02421 , United States
| | - David I Walsh
- MIT Lincoln Laboratory , Lexington , Massachusetts 02421 , United States
| | - Johanna Bobrow
- MIT Lincoln Laboratory , Lexington , Massachusetts 02421 , United States
| | - Kimberly Hamad-Schifferli
- Department of Engineering , University of Massachusetts Boston , Boston , Massachusetts 02125 , United States
| | - David S Kong
- MIT Media Lab , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Todd Thorsen
- MIT Lincoln Laboratory , Lexington , Massachusetts 02421 , United States
| | - Keri Mroszczyk
- MIT Lincoln Laboratory , Lexington , Massachusetts 02421 , United States
| | - Peter A Carr
- MIT Lincoln Laboratory , Lexington , Massachusetts 02421 , United States
- Synthetic Biology Center at MIT , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
16
|
Gong Y, Haque S, Chowdhury P, Cory TJ, Kodidela S, Yallapu MM, Norwood JM, Kumar S. Pharmacokinetics and pharmacodynamics of cytochrome P450 inhibitors for HIV treatment. Expert Opin Drug Metab Toxicol 2019; 15:417-427. [PMID: 30951643 DOI: 10.1080/17425255.2019.1604685] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Drugs used in HIV treatment; all protease inhibitors, some non-nucleoside reverse transcriptase inhibitors, and pharmacoenhancers ritonavir and cobicistat can inhibit cytochrome P450 (CYP) enzymes. CYP inhibition can cause clinically significant drug-drug interactions (DDI), leading to increased drug exposure and potential toxicity. Areas covered: A complete understanding of pharmacodynamics and CYP-mediated DDI is crucial to prevent adverse side effects and to achieve optimal efficacy. We summarized the pharmacodynamics of all the CYP inhibitors used for HIV treatment, followed by a discussion of drug interactions between these CYP inhibitors and other drugs, and a discussion on the effect of CYP polymorphisms. We also discussed the potential advancements in improving the pharmacodynamics of these CYP inhibitors by using nanotechnology strategy. Expert opinion: The drug-interactions in HIV patients receiving ARV drugs are complicated, especially when patients are on CYP inhibitors-based ART regimens. Therefore, evaluation of CYP-mediated drug interactions is necessary prior to prescribing ARV drugs to HIV subjects. To improve the treatment efficacy and minimize DDI, novel approaches such as nanotechnology may be the potential alternative approach. However, further studies with large cohort need to be conducted to provide strong evidence for the use of nano-formulated ARVs to effectively treat HIV patients.
Collapse
Affiliation(s)
- Yuqing Gong
- a Department of Pharmaceutical Sciences , College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Sanjana Haque
- a Department of Pharmaceutical Sciences , College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Pallabita Chowdhury
- a Department of Pharmaceutical Sciences , College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Theodore J Cory
- b Department of Clinical Pharmacy and Translational Science , College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Sunitha Kodidela
- a Department of Pharmaceutical Sciences , College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Murali M Yallapu
- a Department of Pharmaceutical Sciences , College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| | - John M Norwood
- c Department of Infectious Disease , College of Medicine, University of Tennessee Health Science Center , Memphis , TN , USA
| | - Santosh Kumar
- a Department of Pharmaceutical Sciences , College of Pharmacy, University of Tennessee Health Science Center , Memphis , TN , USA
| |
Collapse
|
17
|
Charbe NB, Zacconi FC, Amnerkar N, Ramesh B, Tambuwala MM, Clementi E. Bio-analytical Assay Methods used in Therapeutic Drug Monitoring of Antiretroviral Drugs-A Review. CURRENT DRUG THERAPY 2019; 14:16-57. [DOI: 10.2174/1574885514666181217125550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 09/17/2018] [Accepted: 11/09/2018] [Indexed: 12/12/2022]
Abstract
Background: Several clinical trials, as well as observational statistics, have exhibited that the advantages of antiretroviral [ARV] treatment for humans with Human Immunodeficiency Virus / Acquired Immune Deficiency Syndrome HIV/AIDS exceed their risks. Therapeutic drug monitoring [TDM] plays a key role in optimization of ARV therapy. Determination of ARV’s in plasma, blood cells, and other biological matrices frequently requires separation techniques capable of high effectiveness, specific selectivity and high sensitivity. High-performance liquid chromatography [HPLC] coupled with ultraviolet [UV], Photodiode array detectors [PDA], Mass spectrophotometer [MS] detectors etc. are the important quantitative techniques used for the estimation of pharmaceuticals in biological samples. </P><P> Objective: This review article is aimed to give an extensive outline of different bio-analytical techniques which have been reported for direct quantitation of ARV’s. This article aimed to establish an efficient role played by the TDM in the optimum therapeutic outcome of the ARV treatment. It also focused on establishing the prominent role played by the separation techniques like HPLC and UPLC along with the detectors like UV and Mass in TDM. </P><P> Methods: TDM is based on the principle that for certain drugs, a close relationship exists between the plasma level of the drug and its clinical effect. TDM is of no value if the relationship does not exist. The analytical methodology employed in TDM should: 1) distinguish similar compounds; 2) be sensitive and precise and 3) is easy to use. </P><P> Results: This review highlights the advancement of the chromatographic techniques beginning from the HPLC-UV to the more advanced technique like UPLC-MS/MS. TDM is essential to ensure adherence, observe viral resistance and to personalize ARV dose regimens. It is observed that the analytical methods like immunoassays and liquid chromatography with detectors like UV, PDA, Florescent, MS, MS/MS and Ultra performance liquid chromatography (UPLC)-MS/MS have immensely contributed to the clinical outcome of the ARV therapy. Assay methods are not only helping physicians in limiting the side effects and drug interactions but also assisting in monitoring patient’s compliance. </P><P> Conclusion: The present review revealed that HPLC has been the most widely used system irrespective of the availability of more sensitive chromatographic technique like UPLC.
Collapse
Affiliation(s)
- Nitin B. Charbe
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna McKenna 4860, Macul, Santiago 7820436, Chile
| | - Flavia C. Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna McKenna 4860, Macul, Santiago 7820436, Chile
| | - Nikhil Amnerkar
- Adv V. R. Manohar Institute of Diploma in Pharmacy, Wanadongri, Hingna Road, Nagpur, Maharashtra 441110, India
| | - B. Ramesh
- Sri Adichunchunagiri University, Sri Adichunchunagiri College of Pharmacy, BG Nagar, Karnataka 571418, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, University of Ulster, Coleraine, County Londonderry, Northern Ireland BT52 1SA, United Kingdom
| | - Emilio Clementi
- Clinical Pharmacology Unit, CNR Institute of Neuroscience, Department of Biomedical and Clinical Sciences, Luigi Sacco University Hospital, Universita di Milano, Milan, Italy
| |
Collapse
|
18
|
|
19
|
Lloret-Linares C, Rahmoun Y, Lopes A, Chopin D, Simoneau G, Green A, Delhotal B, Sauvageon H, Mouly S, Bergmann JF, Sellier PO. Effect of body weight and composition on efavirenz, atazanavir or darunavir concentration. Therapie 2018; 73:185-191. [DOI: 10.1016/j.therap.2017.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 07/24/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022]
|
20
|
Deng J, Zhu X, Chen Z, Fan CH, Kwan HS, Wong CH, Shek KY, Zuo Z, Lam TN. A Review of Food–Drug Interactions on Oral Drug Absorption. Drugs 2017; 77:1833-1855. [DOI: 10.1007/s40265-017-0832-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
21
|
Panagopoulos P, Maltezos E, Hatzakis A, Paraskevis D. Hyperbilirubinemia in atazanavir treated HIV-infected patients: the impact of the UGT1A1*28 allele. Pharmgenomics Pers Med 2017; 10:205-208. [PMID: 28790862 PMCID: PMC5488765 DOI: 10.2147/pgpm.s107152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Combination antiretroviral treatment (cART) has significantly improved the life expectancy of people living with HIV. The life-long nature of cART increases the risk of side effects, which in some cases may have been caused by specific genetic characteristics. Patients treated with atazanavir (ATV) boosted with ritonavir (rit), which is a protease inhibitor used for the treatment of HIV, present with elevated bilirubin levels, at high proportions. ATV/rit-related hyperbilirubinemia has been previously associated with genetic characteristics in uridine diphosphate glucuronosyltransferase (UGT) enzyme. The prevalence of the UGT1A1*28 variant, which is the most frequent polymorphism in the UGT1A1 superfamily, has been found to range between 9% and ~60% with the highest frequency in Africa. Pharmacokinetics for additional HIV drugs, such as the integrase inhibitors Raltegravir and Elvitegravir, has been also shown to be influenced by UGT1A1 polymorphisms. Pharmacogenetics/pharmacogenomics testing can be useful to identify a patient's susceptibility to drug toxicity and therefore to facilitate selection of the optimal long-term suppressive regimen.
Collapse
Affiliation(s)
- Periklis Panagopoulos
- 2 Department of Internal Medicine, Democritus University of Thrace, Alexandroupoli, Greece
| | - Efstathios Maltezos
- 2 Department of Internal Medicine, Democritus University of Thrace, Alexandroupoli, Greece
| | - Angelos Hatzakis
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Paraskevis
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Biagi M, Badowski ME, Chiampas T, Young J, Patel M, Vaughn P. Co-administration of elvitegravir/cobicistat/tenofovir disoproxil fumarate/emtricitabine and atazanavir in treatment-experienced HIV patients. Int J STD AIDS 2016; 28:766-772. [PMID: 27587601 DOI: 10.1177/0956462416666440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We report the use of elvitegravir 150 mg/cobicistat 150 mg/tenofovir disoproxil fumarate 300 mg/emtricitabine 200 mg (EVG/COBI/TDF/FTC) once daily, in addition to once-daily atazanavir (ATV) 300 mg, in treatment-experienced patients with human immunodeficiency virus (HIV). Due to limited data available on the co-administration of these agents, our objective was to evaluate and monitor safety and efficacy of this regimen in patients who developed resistance or intolerance to conventional antiretroviral therapy (ART). This short report included offenders incarcerated in the Illinois Department of Corrections who were ≥18 years, HIV-infected, had documented antiretroviral resistance, and received EVG/COBI/TDF/FTC + ATV once daily. Based on previous ART, resistance patterns and current medications, seven patients were initiated on once-daily therapy consisting of EVG/COBI/TDF/FTC and ATV. Due to extensive resistance, two of the seven patients were also started on abacavir (ABC) 600 mg daily in addition to EVG/COBI/TDF/FTC and ATV. Of the seven patients, one had ART changed due to concerns of resistance based on a genotype, one experienced a decline in renal function that warranted a change in therapy, and one is currently virologically suppressed on a combination of EVG/COBI/TDF/FTC, ATV, and ABC. The remaining four patients remain virologically suppressed on EVG/COBI/TDF/FTC + ATV. Therapy consisting of EVG/COBI/TDF/FTC and ATV may be a viable option for some treatment-experienced HIV-infected patients. Further studies evaluating the safety, efficacy, and pharmacokinetics of this therapy are warranted, given the lack of information currently available.
Collapse
Affiliation(s)
- M Biagi
- 1 Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - M E Badowski
- 1 Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - T Chiampas
- 1 Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - J Young
- 2 Section of Infectious Diseases, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - M Patel
- 2 Section of Infectious Diseases, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - P Vaughn
- 3 Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
23
|
Abstract
Drug-interaction issues continue to present a major dilemma for the clinician caring for complex patients such as those infected with HIV. The inherent possibility of a drug interaction is magnified by the multitude of drugs being administered in highly-active antiretroviral therapy (HAART). In addition, other classes of medications are used to alleviate side effects, reduce toxicities associated with HAART, or treat concomitant diseases. The modification of one drug by another substance or drug-drug interaction is the main focus of this article. Drug-drug interactions may result in toxicity, treatment failure, or loss of effectiveness and can significantly affect a patient’s clinical outcome. An understanding of the fundamental mechanisms of HIV drug-drug interactions may allow for the early detection or avoidance of troublesome regimens and prudent management if they develop. Although HIV drug interactions are usually thought of as detrimental, resulting in a loss of therapeutic effect or toxicity, some drug interactions such as ritonavir boosted protease inhibitor–based antiretroviral treatments are beneficial and are commonly used in clinical practice. Therefore, pharmacists need to understand drug interaction mechanisms, remember key drug interactions, and vigilantly monitor patients for potential complications.
Collapse
Affiliation(s)
- Susan A. Krikorian
- Department of Pharmacy Practice, School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences; Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Dorothea C. Rudorf
- Department of Pharmacy Practice, School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences; Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
24
|
Rudorf DC, Krikorian SA. Adverse Effects Associated With Antiretroviral Therapy and Potential Management Strategies. J Pharm Pract 2016. [DOI: 10.1177/0897190005278510] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A variety of adverse drug reactions (ADRs) affecting many organ systems may be observed with antiretroviral therapy (ARV), and they can be differentiated into short- and long- term effects, class effects, or individual drug effects. Commonly seen ADRs include dermatological reactions, associated with nonnucleoside reverse transcriptase inhibitors (NNRTIs) and some protease inhibitors (PIs), and gastrointestinal problems, a major side effect of PIs and of some nucleoside reverse transcriptase inhibitors (NRTIs). Metabolic complications are frequently reported in HIV-infected patients on ARV and often coexist. Lipodystrophy, hyperinsulinemia/hyperglycemia, and bone disorders (osteoporosis, osteonecrosis) are mainly associated with PIs, while lactic acidemia/acidosis are primarily a problem of NRTIs. Hyperlipidemia may be caused by almost all PIs, few NRTIs, and NNRTIs. All antiretroviral drug classes may cause both asymptomatic and symptomatic hepatotoxicity, although nevirapine is the agent most implicated in hepatic events. More drug-specific ADRs include nephrotoxicity (indinavir and tenofovir), central nervous system problems (efavirenz), hematological disturbances (zidovudine), and hypersensitivity reactions (abacavir). Anticipation of ADRs may influence a patient’s decision to delay ARV or to choose specific and potentially less active agents. Occurrence of ADRs may significantly impact a patient’s quality of life and drug adherence. Pharmacists counseling HIV-infected patients should be aware of common ADRs with ARV and potential management strategies.
Collapse
Affiliation(s)
- Dorothea C. Rudorf
- Department of Pharmacy Practice, School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences; Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Susan A. Krikorian
- Department of Pharmacy Practice, School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences; Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
25
|
HIV-1 Alters Intestinal Expression of Drug Transporters and Metabolic Enzymes: Implications for Antiretroviral Drug Disposition. Antimicrob Agents Chemother 2016; 60:2771-81. [PMID: 26902756 DOI: 10.1128/aac.02278-15] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/14/2016] [Indexed: 01/05/2023] Open
Abstract
This study investigated the effects of HIV-1 infection and antiretroviral therapy (ART) on the expression of intestinal drug efflux transporters, i.e., P-glycoprotein (Pgp), multidrug resistance-associated proteins (MRPs), and breast cancer resistance protein (BCRP), and metabolic enzymes, such as cytochrome P450s (CYPs), in the human upper intestinal tract. Intestinal biopsy specimens were obtained from HIV-negative healthy volunteers, ART-naive HIV-positive (HIV(+)) subjects, and HIV(+) subjects receiving ART (10 in each group). Intestinal tissue expression of drug transporters and metabolic enzymes was examined by microarray, real-time quantitative reverse transcription-PCR (qPCR), and immunohistochemistry analyses. Microarray analysis demonstrated significantly lower expression of CYP3A4 and ABCC2/MRP2 in the HIV(+) ART-naive group than in uninfected subjects. qPCR analysis confirmed significantly lower expression of ABCC2/MRP2 in ART-naive subjects than in the control group, while CYP3A4 and ABCG2/BCRP showed a trend toward decreased expression. Protein expression of MRP2 and BCRP was also significantly lower in the HIV(+) naive group than in the control group and was partially restored to baseline levels in HIV(+) subjects receiving ART. In contrast, gene and protein expression of ABCB1/Pgp was significantly increased in HIV(+) subjects on ART relative to HIV(+) ART-naive subjects. These data demonstrate that the expression of drug-metabolizing enzymes and efflux transporters is significantly altered in therapy-naive HIV(+) subjects and in those receiving ART. Since CYP3A4, Pgp, MRPs, and BCRP metabolize or transport many antiretroviral drugs, their altered expression with HIV infection may negatively impact drug pharmacokinetics in HIV(+) subjects. This has clinical implications when using data from healthy volunteers to guide ART.
Collapse
|
26
|
Pasin VP, Pereira AR, Carvalho KAD, Paiva JMGD, Enokihara MMSES, Porro AM. New drugs, new challenges for dermatologists: mucocutaneous ulcers secondary to everolimus. An Bras Dermatol 2016; 90:165-7. [PMID: 26312705 PMCID: PMC4540539 DOI: 10.1590/abd1806-4841.20153672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 05/21/2014] [Indexed: 01/17/2023] Open
Abstract
Everolimus, a mammalian target of rapamycin inhibitor, is an emerging drug, which is
being increasingly applied in oncology and solid organ transplantation. Oral ulcers
are a frequent side effect associated with this immunosupressor. We report the case
of a renal transplant recipient who developed disfiguring oral and perianal ulcers
secondary to everolimus's toxicity. This is probably the first report of perianal
involvement. Dermatologists need to be aware of the potential mucocutaneous adverse
effects related to these new drugs that are becoming evermore common in our clinical
practice.
Collapse
|
27
|
Abstract
Viruses are major pathogenic agents causing a variety of serious diseases in humans, other animals, and plants. Drugs that combat viral infections are called antiviral drugs. There are no effective antiviral drugs for many viral infections. However, there are several drugs for influenza, a couple of drugs for herpesviruses, and some new antiviral drugs for treatment of HIV and hepatitis C infections. The arsenal of antivirals is complex. As of March 2014, it consists of approximately 50 drugs approved by the FDA, approximately half of which are directed against HIV. Antiviral drug creation strategies are focused on two different approaches: targeting the viruses themselves or targeting host cell factors. Direct virus-targeting antiviral drugs include attachment inhibitors, entry inhibitors, uncoating inhibitors, protease inhibitors, polymerase inhibitors, nucleoside and nucleotide reverse transcriptase inhibitors, nonnucleoside reverse-transcriptase inhibitors, and integrase inhibitors. Protease inhibitors (darunavir, atazanavir, and ritonavir), viral DNA polymerase inhibitors (acyclovir, valacyclovir, valganciclovir, and tenofovir), and an integrase inhibitor (raltegravir) are included in the list of Top 200 Drugs by sales for the 2010s.
Collapse
|
28
|
Aceti A, Gianserra L, Lambiase L, Pennica A, Teti E. Pharmacogenetics as a tool to tailor antiretroviral therapy: A review. World J Virol 2015; 4:198-208. [PMID: 26279982 PMCID: PMC4534812 DOI: 10.5501/wjv.v4.i3.198] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 01/20/2015] [Accepted: 07/27/2015] [Indexed: 02/05/2023] Open
Abstract
Highly active antiretroviral therapy (HAART) has substantially changed human immunodeficiency virus (HIV) infection from an inexorably fatal condition into a chronic disease with a longer life expectancy. This means that HIV patients should receive antiretroviral drugs lifelong, and the problems concerning with a chronic treatment (tolerability, side effects, adherence to treatment) have now become dominant. In this context, strategies for the treatment personalization have taken a central role in optimizing the therapeutic response and prevention of adverse drug reactions. In this setting, the study of pharmacogenetics features could be a very useful tool in clinical practice; moreover, nowadays the study of genetic profiles allows optimizations in the therapeutic management of People Living With HIV (PLWH) through the use of test introduced into clinical practice and approved by international guidelines for the adverse effects prevention such as the genetic test HLA-B*5701 to detect hypersensitivity to Abacavir. For other tests further studies are needed: CYP2B6 516 G > T testing may be able to identify patients at higher risk of Central Nervous System side effects following standard dosing of Efavirenz, UGT1A1*28 testing before initiation of antiretroviral therapy containing Atazanavir may aid in identifying individuals at risk of hyperbilirubinaemia. Pharmacogenetics represents a research area with great growth potential which may be useful to guide the rational use of antiretrovirals.
Collapse
|
29
|
Abara WE, Smith L, Zhang S, Fairchild AJ, Heiman HJ, Rust G. The influence of race and comorbidity on the timely initiation of antiretroviral therapy among older persons living with HIV/AIDS. Am J Public Health 2014; 104:e135-41. [PMID: 25211735 DOI: 10.2105/ajph.2014.302227] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVES We examined whether the timely initiation of antiretroviral therapy (ART) differed by race and comorbidity among older (≥ 50 years) people living with HIV/AIDS (PLWHA). METHODS We conducted frequency and descriptive statistics analysis to characterize our sample, which we drew from 2005-2007 Medicaid claims data from 14 states. We employed univariate and multivariable Cox regression analyses to evaluate the relationship between race, comorbidity, and timely ART initiation (≤ 90 days post-HIV/AIDS diagnosis). RESULTS Approximately half of the participants did not commence ART promptly. After we adjusted for covariates, we found that older PLWHA who reported a comorbidity were 40% (95% confidence interval = 0.26, 0.61) as likely to commence ART promptly. We found no racial differences in the timely initiation of ART among older PLWHA. CONCLUSIONS Comorbidities affect timely ART initiation in older PLWHA. Older PLWHA may benefit from integrating and coordinating HIV care with care for other comorbidities and the development of ART treatment guidelines specific to older PLWHA. Consistent Medicaid coverage helps ensure consistent access to HIV treatment and care and may eliminate racial disparities in timely ART initiation among older PLWHA.
Collapse
Affiliation(s)
- Winston E Abara
- Winston E. Abara is with the Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA. Lerissa Smith and Harry J. Heiman are with the Satcher Health Leadership Institute, Morehouse School of Medicine, Atlanta. Shun Zhang and George Rust are with the National Center for Primary Care, Morehouse School of Medicine. Amanda J. Fairchild is with the Department of Psychology, University of South Carolina, Columbia
| | | | | | | | | | | |
Collapse
|
30
|
Kis O, Walmsley SL, Bendayan R. In Vitro and In Situ Evaluation of pH-Dependence of Atazanavir Intestinal Permeability and Interactions with Acid-Reducing Agents. Pharm Res 2014; 31:2404-19. [DOI: 10.1007/s11095-014-1336-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 02/08/2014] [Indexed: 12/12/2022]
|
31
|
Lozano R, Domeque N, Apesteguia AF. Extended mathematical model for "in vivo" quantification of the interaction betweeen atazanavir and bilirubin. J Clin Pharmacol 2013; 54:161-7. [PMID: 24243081 DOI: 10.1002/jcph.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 11/13/2013] [Indexed: 11/07/2022]
Abstract
The objective of the present work was to conduct an "in vivo" analysis of the atazanavir-bilirubin interaction. We developed a new mathematical approach to PK/PDPK models for competitive interaction based on the Michaelis-Menten equation, which was applied to patients with polymorphisms in the gene for UDP-glucuronosyltransferase 1A1 (UGT1A1). Atazanavir is known to induce concentration-dependent increases in bilirubin plasma levels. Thus, we employed our mathematical model to analyse rises in steady state atazanavir and bilirubin concentrations, ultimately plotting a nomogram for detection of suboptimal atazanavir exposure. Application of our model revealed that an absolute value or a steady state increase in bilirubin falling below 3.8Φ µmol/L (where Φ is a correction factor, =1 for UGT1A1 wild type and ≠1 for UGT1A1 variants) could be used to predict suboptimal atazanavir exposure and treatment failure. Thus, we have successfully established a new mathematical approach for pharmacodynamic-pharmacokinetic modelling of the interaction between atazanavir and bilirubin, as it relates to genetic variants of UGT1A1. Taken together, our findings indicate that bilirubin plasma levels represent a valuable marker of atazanavir exposure.
Collapse
Affiliation(s)
- Roberto Lozano
- Department of Pharmacy, Hospital Real Ntra. Sra. de Gracia, Zaragoza, Spain
| | | | | |
Collapse
|
32
|
Lozano R, Domeque N, Apesteguia AF. Atazanavir-bilirubin interaction: a pharmacokinetic-pharmacodynamic model. Clin Pharmacol 2013; 5:153-9. [PMID: 24106429 PMCID: PMC3792011 DOI: 10.2147/cpaa.s48377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Purpose The aim of this work was to analyze the atazanavir–bilirubin relationship, using a new mathematical approach to pharmacokinetic–pharmacodynamic models, for competitive drug interactions based on Michaelis–Menten equations. Patients and methods Because atazanavir induces an increase of plasma bilirubin levels, in a concentration-dependent manner, we developed a mathematical model, based on increments of atazanavir and bilirubin concentrations at steady state, in HIV infected (HIV+) patients, and plotted the corresponding nomogram for detecting suboptimal atazanavir exposure. Results By applying the obtained model, the results indicate that an absolute value or an increment of bilirubin at steady state below 3.8 μmol/L, are predictive of suboptimal atazanavir exposure and therapeutic failure. Conclusion We have successfully implemented a new mathematical approach to pharmacokinetic–pharmacodynamic model for atazanavir–bilirubin interaction. As a result, we found that bilirubin plasma levels constitute a good marker of exposure to atazanavir and of viral suppression.
Collapse
Affiliation(s)
- Roberto Lozano
- Pharmacy Department, Hospital Real Nuestra, Señora de Gracia
| | | | | |
Collapse
|
33
|
Zhu L, Persson A, Mahnke L, Eley T, Li T, Xu X, Agarwala S, Dragone J, Bertz R. Effect of Low-Dose Omeprazole (20 mg Daily) on the Pharmacokinetics of Multiple-Dose Atazanavir With Ritonavir in Healthy Subjects. J Clin Pharmacol 2013; 51:368-77. [DOI: 10.1177/0091270010367651] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Reese MJ, Savina PM, Generaux GT, Tracey H, Humphreys JE, Kanaoka E, Webster LO, Harmon KA, Clarke JD, Polli JW. In vitro investigations into the roles of drug transporters and metabolizing enzymes in the disposition and drug interactions of dolutegravir, a HIV integrase inhibitor. Drug Metab Dispos 2013; 41:353-61. [PMID: 23132334 DOI: 10.1124/dmd.112.048918] [Citation(s) in RCA: 210] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Dolutegravir (DTG; S/GSK1349572) is a potent HIV-1 integrase inhibitor with a distinct resistance profile and a once-daily dose regimen that does not require pharmacokinetic boosting. This work investigated the in vitro drug transport and metabolism of DTG and assessed the potential for clinical drug-drug interactions. DTG is a substrate for the efflux transporters P-glycoprotein (Pgp) and human breast cancer resistance protein (BCRP). Its high intrinsic membrane permeability limits the impact these transporters have on DTG's intestinal absorption. UDP-glucuronosyltransferase (UGT) 1A1 is the main enzyme responsible for the metabolism of DTG in vivo, with cytochrome P450 (P450) 3A4 being a notable pathway and UGT1A3 and UGT1A9 being only minor pathways. DTG demonstrated little or no inhibition (IC(50) values > 30 μM) in vitro of the transporters Pgp, BCRP, multidrug resistance protein 2, organic anion transporting polypeptide 1B1/3, organic cation transporter (OCT) 1, or the drug metabolizing enzymes CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 3A4, UGT1A1, or 2B7. Further, DTG did not induce CYP1A2, 2B6, or 3A4 mRNA in vitro using human hepatocytes. DTG does inhibit the renal OCT2 (IC(50) = 1.9 μM) transporter, which provides a mechanistic basis for the mild increases in serum creatinine observed in clinical studies. These in vitro studies demonstrate a low propensity for DTG to be a perpetrator of clinical drug interactions and provide a basis for predicting when other drugs could result in a drug interaction with DTG.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2
- ATP-Binding Cassette Transporters/antagonists & inhibitors
- ATP-Binding Cassette Transporters/metabolism
- Animals
- CHO Cells
- Cricetinae
- Cricetulus
- Cytochrome P-450 Enzyme Inhibitors
- Cytochrome P-450 Enzyme System/genetics
- Cytochrome P-450 Enzyme System/metabolism
- Dogs
- Drug Interactions
- Enzyme Induction
- Female
- Glucuronosyltransferase/antagonists & inhibitors
- Glucuronosyltransferase/genetics
- Glucuronosyltransferase/metabolism
- HIV Integrase Inhibitors/metabolism
- HIV Integrase Inhibitors/pharmacology
- Hepatocytes/drug effects
- Hepatocytes/enzymology
- Heterocyclic Compounds, 3-Ring/metabolism
- Heterocyclic Compounds, 3-Ring/pharmacology
- Humans
- Isoenzymes
- Madin Darby Canine Kidney Cells
- Male
- Membrane Transport Proteins/drug effects
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/metabolism
- Microsomes, Liver/drug effects
- Microsomes, Liver/enzymology
- Multidrug Resistance-Associated Protein 2
- Multidrug Resistance-Associated Proteins/antagonists & inhibitors
- Multidrug Resistance-Associated Proteins/metabolism
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Organic Anion Transporters/antagonists & inhibitors
- Organic Anion Transporters/metabolism
- Oxazines
- Piperazines
- Pyridones
- Transfection
Collapse
Affiliation(s)
- Melinda J Reese
- Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Although the past 20 years have seen many advances in HIV pharmacotherapy, host variability has only been more recently recognized as an important driver of both treatment response and toxicity. This review will focus on the importance of variability in drug metabolizing enzymes and the potential research and clinical applications of these findings in HIV care. RECENT FINDINGS The treatment of HIV is complex and involves chronic treatment with multiple drugs and drug classes. Recent research has been important in uncovering genetic differences in drug metabolizing and drug transporter genes, which underpin dose-dependent toxicity and efficacy, but also in genes defining differences in immune response and human leukocyte antigen genes, which restrict processes that are less dependent on the drug dose. SUMMARY The advancement of pharmacogenetics will continue to further our knowledge of disease-drug interactions and pathogenesis. Ultimately, the goals of pharmacogenetics and pharmacogenomics in HIV medicine will be the development of the right drugs for the right patient or population. The strong association between HLA-B5701 and abacavir hypersensitivity reaction shows promise for a genetic screening test to be feasibly incorporated into clinical practice. The clinical applicability of many findings demonstrating associations between single nucleotide polymorphisms, particularly in drug metabolism and transporter genes and drug efficacy and toxicity, are currently uncertain.
Collapse
|
36
|
Michaud V, Bar-Magen T, Turgeon J, Flockhart D, Desta Z, Wainberg MA. The dual role of pharmacogenetics in HIV treatment: mutations and polymorphisms regulating antiretroviral drug resistance and disposition. Pharmacol Rev 2012; 64:803-33. [PMID: 22759796 DOI: 10.1124/pr.111.005553] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Significant intra- and interindividual variability has been observed in response to use of pharmacological agents in treatment of HIV infection. Treatment of HIV infection is limited by high rates of adverse drug reactions and development of resistance in a significant proportion of patients as a result of suboptimal drug concentrations. The efficacy of antiretroviral therapy is challenged by the emergence of resistant HIV-1 mutants with reduced susceptibility to antiretroviral drugs. Moreover, pharmacotherapy of patients infected with HIV is challenging because a great number of comorbidities increase polypharmacy and the risk for drug-drug interactions. Drug-metabolizing enzymes and drug transporters regulate drug access to the systemic circulation, target cells, and sanctuary sites. These factors, which determine drug exposure, along with the emergence of mutations conferring resistance to HIV medications, could explain variability in efficacy and adverse drug reactions associated with antiretroviral drugs. In this review, the major factors affecting the disposition of antiretroviral drugs, including key drug-metabolizing enzymes and membrane drug transporters, are outlined. Genetic polymorphisms affecting the activity and/or the expression of cytochromes P450 or UGT isozymes and membrane drug transport proteins are highlighted and include such examples as the association of neurotoxicity with efavirenz, nephrotoxicity with tenofovir, hepatotoxicity with nevirapine, and hyperbilirubinemia with indinavir and atazanavir. Mechanisms of drug resistance conferred by specific viral mutations are also reviewed, with particular attention to replicative viral fitness and transmitted HIV drug resistance with the objectives of providing a better understanding of mechanisms involved in HIV drug resistance and helping health care providers to better manage interpatient variability in drug efficacy and toxicity.
Collapse
Affiliation(s)
- Veronique Michaud
- McGill University AIDS Centre, Lady Davis Institute for Medical Research, 3755 Cote-Ste-Catherine Rd., Montréal, Québec, H3T 1E2, Canada
| | | | | | | | | | | |
Collapse
|
37
|
No effect of a single supratherapeutic dose of lersivirine, a next-generation nonnucleoside reverse transcriptase inhibitor, on corrected QT interval in healthy subjects. Antimicrob Agents Chemother 2012; 56:2408-13. [PMID: 22371898 DOI: 10.1128/aac.05194-11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The objective of this study was to investigate the effect of a supratherapeutic dose of lersivirine (LRV) on corrected QT (QTc) interval using Fridericia's equation (QTcF) in healthy subjects. In this randomized, single-dose, placebo- and active-controlled 3-way crossover study, healthy adult males (n = 48) were randomized to receive LRV (2,400 mg), moxifloxacin (400 mg), or placebo for each treatment period. Triplicate 12-lead electrocardiogram measurements were performed, PK samples were collected, and vital signs were measured. Adverse event monitoring and safety laboratory testing were performed. All subjects were white (mean age, 39 years; body mass index [BMI], 25.6 kg/m(2)) and completed the study. Following LRV administration, the upper bound of the 90% confidence interval (CI) for time-matched adjusted mean differences to placebo QTcF at each time point postdose was below the regulatory threshold of 10 ms, satisfying the criteria for a negative thorough QT/QTc study. The highest upper bound of QTcF 90% CI occurred at 6 h for LRV (3.32 ms; 90% CI, 1.47 to 5.17 ms). The study was deemed adequately sensitive as the lower bound of the 90% CI for the adjusted mean QTcF differences between moxifloxacin and placebo at the moxifloxacin historical T(max) of 3 h was >5 ms (15.29 ms; 90% CI, 13.44 to 17.14 ms). There was no statistically significant relationship between LRV exposure and placebo-adjusted change from baseline QTcF or clinically significant changes in QRS complex, pulse rate (PR) interval, heart rate, or blood pressure. LRV (2,400 mg) did not prolong the QTcF interval, and no clinically relevant electrocardiogram or vital sign changes were observed in healthy subjects.
Collapse
|
38
|
Rekić D, Clewe O, Röshammar D, Flamholc L, Sönnerborg A, Ormaasen V, Gisslén M, Abelö A, Ashton M. Bilirubin-a potential marker of drug exposure in atazanavir-based antiretroviral therapy. AAPS JOURNAL 2011; 13:598-605. [PMID: 21913053 DOI: 10.1208/s12248-011-9299-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/24/2011] [Indexed: 01/11/2023]
Abstract
The objective of this work was to examine the atazanavir-bilirubin relationship using a population-based approach and to assess the possible application of bilirubin as a readily available marker of atazanavir exposure. A model of atazanavir exposure and its concentration-dependent effect on bilirubin levels was developed based on 200 atazanavir and 361 bilirubin samples from 82 patients receiving atazanavir in the NORTHIV trial. The pharmacokinetics was adequately described by a one-compartment model with first-order absorption and lag-time. The maximum inhibition of bilirubin elimination rate constant (I(max)) was estimated at 91% (95% CI, 87-94) and the atazanavir concentration resulting in half of I(max) (IC50) was 0.30 μmol/L (95% CI, 0.24-0.37). At an atazanavir/ritonavir dose of 300/100 mg given once daily, the bilirubin half-life was on average increased from 1.6 to 8.1 h. A nomogram, which can be used to indicate suboptimal atazanavir exposure and non-adherence, was constructed based on model simulations.
Collapse
Affiliation(s)
- Dinko Rekić
- Unit for Pharmacokinetics and Drug Metabolism, Department of Pharmacology, Sahlgrenska Academy at University of Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Karlgren M, Ahlin G, Bergström CAS, Svensson R, Palm J, Artursson P. In vitro and in silico strategies to identify OATP1B1 inhibitors and predict clinical drug-drug interactions. Pharm Res 2011; 29:411-26. [PMID: 21861202 PMCID: PMC3264873 DOI: 10.1007/s11095-011-0564-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 08/08/2011] [Indexed: 12/02/2022]
Abstract
Purpose To establish in vitro and in silico models that predict clinical drug–drug interactions (DDIs) with the OATP1B1 (SLCO1B1) transporter. Methods The inhibitory effect of 146 drugs and drug-like compounds on OATP1B1-mediated transport was studied in HEK293 cells. A computational model was developed to predict OATP1B1 inhibition. Concentration-dependent effects were investigated for six compounds; clinical DDIs were predicted by calculating change in exposure (i.e. R-values) in eight different ways. Results Sixty-five compounds were identified as OATP1B1 inhibitors at 20 μM. The computational model predicted the test set with 80% accuracy for inhibitors and 91% for non-inhibitors. In vitro–in vivo comparisons underscored the importance of using drugs with known clinical effects as references. Thus, reference drugs, cyclosporin A, gemfibrozil, and fenofibrate, provided an inhibition interval to which three antiviral drugs, atazanavir, lopinavir, and amprenavir, could be compared and their clinical DDIs with OATP1B1 classified. Conclusions Twenty-two new OATP1B1 inhibitors were identified, a predictive OATP1B1 inhibition in silico model was developed, and successful predictions of clinical DDIs were obtained with OATP1B1.
Collapse
Affiliation(s)
- Maria Karlgren
- Uppsala University Drug Optimization and Pharmaceutical Profiling Platform, Department of Pharmacy, Uppsala University, The Biomedical Centre, P.O. Box 580, 751 23, Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
40
|
Severe skin rash associated with atazanavir. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2011; 20:e10-2. [PMID: 20190882 DOI: 10.1155/2009/721956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Accepted: 09/26/2008] [Indexed: 01/11/2023]
Abstract
Three cases of severe rash associated with the use of atazanavir are described. In all cases, the rash was maculopapular and pruritic. Rash onset occurred eight to 11 days after initiation of therapy, and resolved with atazanavir discontinuation. Clinicians prescribing atazanavir should be aware of this potential adverse effect.
Collapse
|
41
|
Pharmacokinetics of once-daily darunavir-ritonavir and atazanavir-ritonavir over 72 hours following drug cessation. Antimicrob Agents Chemother 2011; 55:4218-23. [PMID: 21709075 DOI: 10.1128/aac.01747-10] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The object of this study was to investigate the pharmacokinetics of darunavir-ritonavir and atazanavir-ritonavir once-daily dosing over 72 h (h) following drug intake cessation. Volunteers received darunavir-ritonavir at 800 and 100 mg, respectively, once daily for 10 days, followed by a 7-day washout period, and atazanavir-ritonavir at 300 and 100 mg, respectively, once daily for 10 days. Full pharmacokinetic profiles were assessed for each phase for the 72 h following day 10. Pharmacokinetic parameters were determined over 24 h and to the last measurable concentration by noncompartmental methods. Seventeen subjects completed the study. The geometric mean (GM) terminal elimination half-life to 72 h of darunavir was 6.48 h, which was lower than the 0- to 24-h half-life (10.70 h). The terminal elimination half-life of atazanavir was 6.74 h, which was lower than the 0- to 24-h half-life (13.72 h). All subjects but one had darunavir concentrations higher than the target of 550 ng/ml for protease-resistant HIV isolates (equivalent to 10 times the protein-binding-corrected 50% inhibitory concentration [IC(50)] for wild-type virus) at 24 h postdose, and 14 out of 17 had concentrations higher than the target at 30 h postdose (GM of 1,088 and 851 ng/ml). All subjects had atazanavir concentrations above the suggested minimum effective concentration of 150 ng/ml (equivalent to 10 times the protein-binding-corrected IC(50) for wild-type virus) at 24 and 30 h postdose (GM of 693 and 392 ng/ml). Two of 17 and 5 of 17 subjects were above target at 48 h postdose while on darunavir-ritonavir and atazanavir-ritonavir. Ritonavir half-life to 72 h was 6.84 h with darunavir and 6.07 with atazanavir. This study investigated the pharmacokinetic forgiveness of two boosted protease inhibitors. Although the rates of decline of darunavir and atazanavir slightly increased as ritonavir concentrations declined, most individuals had concentrations 6 h after the end of the ideal dosing interval of 24 h which were above the cutoff used to define therapeutic concentrations.
Collapse
|
42
|
Solas C, Muret P. Niveau de preuve du suivi therapeutique pharmacologique de l’atazanavir. Therapie 2011; 66:213-9. [DOI: 10.2515/therapie/2011032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 01/03/2011] [Indexed: 01/11/2023]
|
43
|
Neely MN, Rakhmanina NY. Pharmacokinetic Optimization of Antiretroviral Therapy in Children and Adolescents. Clin Pharmacokinet 2011; 50:143-89. [DOI: 10.2165/11539260-000000000-00000] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
44
|
Li F, Lu J, Wang L, Ma X. CYP3A-mediated generation of aldehyde and hydrazine in atazanavir metabolism. Drug Metab Dispos 2011; 39:394-401. [PMID: 21148252 PMCID: PMC3061559 DOI: 10.1124/dmd.110.036327] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 12/09/2010] [Indexed: 11/22/2022] Open
Abstract
Atazanavir (ATV) is an antiretroviral drug of the protease inhibitor class. Multiple adverse effects of ATV have been reported in clinical practice, such as jaundice, nausea, abdominal pain, and headache. The exact mechanisms of ATV-related adverse effects are unknown. It is generally accepted that a predominant pathway of drug-induced toxicity is through the generation of reactive metabolites. Our current study was designed to explore reactive metabolites of ATV. We used a metabolomic approach to profile ATV metabolism in mice and human liver microsomes. We identified 5 known and 13 novel ATV metabolites. Three potential reactive metabolites were detected and characterized for the first time: one aromatic aldehyde, one α-hydroxyaldehyde, and one hydrazine. These potential reactive metabolites were primarily generated by CYP3A. Our results provide a clue for studies on ATV-related adverse effects from the aspect of metabolic activation. Further studies are suggested to illustrate the impact of these potential reactive metabolites on ATV-related adverse effects.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
45
|
Achenbach CJ, Darin KM, Murphy RL, Katlama C. Atazanavir/ritonavir-based combination antiretroviral therapy for treatment of HIV-1 infection in adults. Future Virol 2011; 6:157-177. [PMID: 21731578 DOI: 10.2217/fvl.10.89] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the past 15 years, improvements in the management of HIV infection have dramatically reduced morbidity and mortality. Similarly, rapid advances in antiretroviral medications have resulted in the possibility of life-long therapy with simple and tolerable regimens. Protease inhibitors have been important medications in regimens of combination antiretroviral therapy for the treatment of HIV. One of the recommended and commonly used therapies in this class is once-daily-administered atazanavir, pharmacologically boosted with ritonavir (atazanavir/r). Clinical studies and practice have shown these drugs, in combination with other antiretroviral agents, to be potent, safe and easy to use in a variety of settings. Atazanavir/r has minimal short-term toxicity, including benign bilirubin elevation, and has less potential for long-term complications of hyperlipidemia and insulin resistance compared with other protease inhibitors. A high genetic barrier to resistance and a favorable resistance profile make it an excellent option for initial HIV treatment or as the first drug utilized in the protease inhibitors class. Atazanavir/r is also currently being studied in novel treatment strategies, including combinations with new classes of antiretrovirals to assess nucleoside reverse transcriptase inhibitor-sparing regimens. In this article we review atazanavir/r as a treatment for HIV infection and discuss the latest information on its pharmacology, efficacy and toxicity.
Collapse
Affiliation(s)
- Chad J Achenbach
- Feinberg School of Medicine & Center for Global Health, Northwestern University, Chicago, USA
| | | | | | | |
Collapse
|
46
|
Müller AC, Kanfer I. An efficient HPLC method for the quantitative determination of atazanavir in human plasma suitable for bioequivalence and pharmacokinetic studies in healthy human subjects. J Pharm Biomed Anal 2010; 53:113-8. [DOI: 10.1016/j.jpba.2010.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 03/15/2010] [Accepted: 03/17/2010] [Indexed: 10/19/2022]
|
47
|
Morillo Verdugo R, Fernández Lisón LC, Huertas Fernández MJ, Martín Conde MT, Roldan Morales JC, Ruano Camps R, Serrano López De Las Hazas JI, Ibarra Barrueta O, Illaro Uranga A. [The role of the hospital pharmacist in the prevention, treatment and management of the side effects associated with antiretroviral treatment]. FARMACIA HOSPITALARIA 2010; 34:237-50. [PMID: 20655783 DOI: 10.1016/j.farma.2010.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 01/20/2010] [Indexed: 11/29/2022] Open
Abstract
At present, the side effects associated with antiretroviral treatment are the main reasons for discontinuation of this kind of therapy, both in clinical trials and in regular clinical practise. On the other hand, due to the change of direction that our profession has suffered in recent years, we face the need to establish a different relationship with the patient, achieving direct and effective Pharmaceutical Care within a framework of shared responsibility for therapeutic results. Pharmacist interventions should be aimed at improving the quality of life of patients, which can only be achieved with a multidisciplinary approach and individualised and adjusted to new patterns of toxicity of the drugs currently used. The pharmacist who does this work must know how to interpret these side effects, giving accurate information to the patient about both pharmacological and non-pharmacological treatment and correct pharmaceutical follow-up which clearly sets forth the criteria for referral to medical appointments. The aim of this paper is to establish baselines so that the hospital pharmacist can perform clearly and uniformly in the prevention, identification and management of major side effects: gastrointestinal, cardiovascular, dermatological, at the central nervous system and kidney level, associated with antiretroviral therapy.
Collapse
Affiliation(s)
- R Morillo Verdugo
- Unidad de Gestión Clínica de Farmacia, Hospital Universitario de Valme, Sevilla, España.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Balayssac E, Autret-Leca E, Jonville-Béra AP, Diè-Kacou H, Beau-Salinas F. [Adverse reactions of atazanavir, fosamprenavir and tipranavir in "real life"]. Therapie 2010; 65:121-8. [PMID: 20478244 DOI: 10.2515/therapie/2010003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 12/08/2009] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To precise adverse effects of atazanavir, fosamprenavir and tipranavir "in real life". METHOD Descriptive study of 3 protease inhibitor adverse effects stored in the French Bank of Pharmacovigilance. RESULTS Nineteen adverse effects having at least possible links with antiretroviral drugs studied were reported. It was essentially hepatobiliary (atazanavir: 29/59, tipranavir: 4/6) and skin (fosamprenavir: 10/20) adverse reactions. These reactions, relatively "serious" (35.1%) led to the interruption of the person (or persons) medication (s) suspected (s) in 69 folds (82.1%) and evolved to healing without sequelae in 68 folds (81%). CONCLUSION The drug side effects were for the most expected. However, their frequency and their seriously underline the interest of a post-AMM monitoring to reassess the drugs risk-benefit report.
Collapse
Affiliation(s)
- Eric Balayssac
- Centre Hospitalier Universitaire de Cocody (Abidjan), Service de Pharmacologie Clinique, Abidjan, Côte d'Ivoire.
| | | | | | | | | |
Collapse
|
49
|
Influence of atazanavir on the pharmacodynamics and pharmacokinetics of gliclazide in animal models. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/j.ijdm.2009.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
50
|
Janneh O, Bray PG, Jones E, Wyen C, Chiba P, Back DJ, Khoo SH. Concentration-dependent effects and intracellular accumulation of HIV protease inhibitors in cultured CD4 T cells and primary human lymphocytes. J Antimicrob Chemother 2010; 65:906-16. [PMID: 20237075 DOI: 10.1093/jac/dkq082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The intracellular and plasma concentrations of HIV protease inhibitors (HPIs) vary widely in vivo. It is unclear whether there is a concentration-dependent effect of HPIs such that at increasing concentration they may either block their own efflux (leading to 'autoboosting') or influx (leading to saturability/decreased intracellular accumulation). METHOD The effects of various concentrations (0-30 microM) of lopinavir, saquinavir, ritonavir and atazanavir on the accumulation of [(14)C]lopinavir, [(3)H]saquinavir, [(3)H]ritonavir and [(3)H]atazanavir, respectively, were investigated in CEM(parental), CEM(VBL) [P-glycoprotein (ABCB1) overexpressing], CEM(E1000) (MRP1 overexpressing) and in peripheral blood mononuclear cells (PBMCs). We also investigated the effects of inhibitors of ABCB1/ABCG2 (tariquidar), ABCC (MK571) and ABCC1/2 (frusemide), singly and in combination with HPIs, on cellular accumulation. RESULTS In all the cell lines, with increasing concentration of lopinavir, saquinavir and ritonavir, there was a significant increase in the cellular accumulation of [(14)C]lopinavir, [(3)H]saquinavir and [(3)H]ritonavir. Tariquidar, MK571 and frusemide (alone and in combination with lopinavir, saquinavir and ritonavir) significantly increased the accumulation of [(14)C]lopinavir, [(3)H]saquinavir and [(3)H]ritonavir. Ritonavir (alone or in combination with tariquidar) decreased the intracellular accumulation of [(3)H]ritonavir in PBMCs. Atazanavir decreased the accumulation of [(3)H]atazanavir in a concentration-dependent manner in all of the cells tested. CONCLUSIONS There are complex and variable drug-specific rather than class-specific effects of the HPIs on their own accumulation.
Collapse
Affiliation(s)
- Omar Janneh
- Department of Biomolecular and Sport Sciences, James Starley Building, Priory Street, Coventry University, Coventry CV1 5FB, UK
| | | | | | | | | | | | | |
Collapse
|