1
|
Zhang P, Qian Y, Wang L, Suo J, Yin L, Wang Y, Zhang L, Wang Z. The Association of Intraocular Efavirenz Concentrations and HIV-1 Viral Load Among Persons With HIV. J Acquir Immune Defic Syndr 2024; 96:299-303. [PMID: 38534141 DOI: 10.1097/qai.0000000000003426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
OBJECTIVE Efavirenz (EFV) is commonly used in combination antiretroviral therapy. However, in our previous study, many persons living with HIV exhibited ocular complications despite undergoing effective combination antiretroviral therapy. Here, we aimed to determine the intraocular EFV concentrations in the vitreous and analyze the factors affecting viral load in the vitreous in patients with HIV-associated retinopathies. DESIGN Observational, retrospective study. METHODS Fourteen patients receiving EFV in combination with an antiretroviral therapy who underwent pars plana vitrectomy were enrolled between January 2019 and August 2022. The patients were divided into 2 groups based on presence or absence of retinal detachment (RD). Patient characteristics and HIV-1 RNA levels in plasma and vitreous were recorded during pars plana vitrectomy. Paired blood plasma and vitreous samples were obtained for EFV concentration analysis using ultra-high-performance liquid chromatography/tandem mass spectrometry. RESULTS The median age of the enrolled patients was 48 years (interquartile range, 32.25-53.25), including 12 men and 2 women. Median vitreous and plasma EFV concentrations were 141.5 (interquartile range, 69.63-323.75) and 2620 ng/mL (1680-4207.5), respectively. Median ratio of vitreous/plasma EFV concentrations in the paired samples among all participants was 0.053 (0.018-0.118). Median vitreous/plasma EFV concentrations significantly differed between the non-RD and RD groups (0.04 vs 0.12, P = 0.042). CONCLUSIONS The vitreous EFV concentrations were insufficient to inhibit viral replication in intraocular tissues, which may be because of poor penetration of the blood-retinal barrier. High vitreous EFV concentrations were associated with RD, indicating a correlation between the EFV concentration and the severity of blood-retinal barrier disruption. It implied that EFV was not a suitable antiviral drug to inhibit HIV-1 replication in ocular tissues.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China ; and
| | - Yiwen Qian
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China ; and
| | - Luoziyi Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China ; and
| | - Jinshan Suo
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China ; and
| | - Lin Yin
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University
| | - Yuceng Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China ; and
| | - Lijun Zhang
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University
| | - Zhiliang Wang
- Department of Ophthalmology, Huashan Hospital of Fudan University, Shanghai, China ; and
| |
Collapse
|
2
|
Mast N, Li Y, Pikuleva IA. 7,8-Dihydroxy Efavirenz Is Not as Effective in CYP46A1 Activation In Vivo as Efavirenz or Its 8,14-Dihydroxy Metabolite. Int J Mol Sci 2024; 25:2242. [PMID: 38396919 PMCID: PMC10889178 DOI: 10.3390/ijms25042242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
High dose (S)-efavirenz (EFV) inhibits the HIV reverse transcriptase enzyme and is used to lower HIV load. Low-dose EFV allosterically activates CYP46A1, the key enzyme for cholesterol elimination from the brain, and is investigated as a potential treatment for Alzheimer's disease. Simultaneously, we evaluate EFV dihydroxymetabolites for in vivo brain effects to compare with those of (S)-EFV. We have already tested (rac)-8,14dihydroxy EFV on 5XFAD mice, a model of Alzheimer's disease. Herein, we treated 5XFAD mice with (rac)-7,8dihydroxy EFV. In both sexes, the treatment modestly activated CYP46A1 in the brain and increased brain content of acetyl-CoA and acetylcholine. Male mice also showed a decrease in the brain levels of insoluble amyloid β40 peptides. However, the treatment had no effect on animal performance in different memory tasks. Thus, the overall brain effects of (rac)-7,8dihydroxy EFV were weaker than those of EFV and (rac)-8,14dihydroxy EFV and did not lead to cognitive improvements as were seen in treatments with EFV and (rac)-8,14dihydroxy EFV. An in vitro study assessing CYP46A1 activation in co-incubations with EFV and (rac)-7,8dihydroxy EFV or (rac)-8,14dihydroxy EFV was carried out and provided insight into the compound doses and ratios that could be used for in vivo co-treatments with EFV and its dihydroxymetabolite.
Collapse
Affiliation(s)
| | | | - Irina A. Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH 44106, USA; (N.M.); (Y.L.)
| |
Collapse
|
3
|
Wheeler AM, Orsburn BC, Bumpus NN. Biotransformation of Efavirenz and Proteomic Analysis of Cytochrome P450s and UDP-Glucuronosyltransferases in Mouse, Macaque, and Human Brain-Derived In Vitro Systems. Drug Metab Dispos 2023; 51:521-531. [PMID: 36623884 PMCID: PMC10043944 DOI: 10.1124/dmd.122.001195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Antiretroviral drugs such as efavirenz (EFV) are essential to combat human immunodeficiency virus (HIV) infection in the brain, but little is known about how these drugs are metabolized locally. In this study, the cytochrome P450 (P450) and UDP-glucuronosyltransferase (UGT)-dependent metabolism of EFV was probed in brain microsomes from mice, cynomolgus macaques, and humans as well as primary neural cells from C57BL/6N mice. Utilizing ultra high performance liquid chromatography high-resolution mass spectrometry (uHPLC-HRMS), the formation of 8-hydroxyefavirenz (8-OHEFV) from EFV and the glucuronidation of P450-dependent metabolites 8-OHEFV and 8,14-dihydroxyefavirenz (8,14-diOHEFV) were observed in brain microsomes from all three species. The direct glucuronidation of EFV, however, was only detected in cynomolgus macaque brain microsomes. In primary neural cells treated with EFV, microglia were the only cell type to exhibit metabolism, forming 8-OHEFV only. In cells treated with the P450-dependent metabolites of EFV, glucuronidation was detected only in cortical neurons and astrocytes, revealing that certain aspects of EFV metabolism are cell type specific. Untargeted and targeted proteomics experiments were used to identify the P450s and UGTs present in brain microsomes. Eleven P450s and 11 UGTs were detected in human brain microsomes, whereas seven P450s and 14 UGTs were identified in mouse brain microsomes and 15 P450s and four UGTs, respectively, were observed in macaque brain microsomes. This was the first time many of these enzymes have been noted in brain microsomes at the protein level. This study indicates the potential for brain metabolism to contribute to pharmacological and toxicological outcomes of EFV in the brain. SIGNIFICANCE STATEMENT: Metabolism in the brain is understudied, and the persistence of human immunodeficiency virus (HIV) infection in the brain warrants the evaluation of how antiretroviral drugs such as efavirenz are metabolized in the brain. Using brain microsomes, the metabolism of efavirenz by both cytochrome P450s (P450s) and UDP-glucuronosyltransferases (UGTs) is established. Additionally, proteomics of brain microsomes characterizes P450s and UGTs in the brain, many of which have not yet been noted in the literature at the protein level.
Collapse
Affiliation(s)
- Abigail M Wheeler
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Benjamin C Orsburn
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Namandjé N Bumpus
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Angle ED, Cox PM. Multidisciplinary Insights into the Structure-Function Relationship of the CYP2B6 Active Site. Drug Metab Dispos 2023; 51:369-384. [PMID: 36418184 DOI: 10.1124/dmd.122.000853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022] Open
Abstract
Cytochrome P450 2B6 (CYP2B6) is a highly polymorphic human enzyme involved in the metabolism of many clinically relevant drugs, environmental toxins, and endogenous molecules with disparate structures. Over the last 20-plus years, in silico and in vitro studies of CYP2B6 using various ligands have provided foundational information regarding the substrate specificity and structure-function relationship of this enzyme. Approaches such as homology modeling, X-ray crystallography, molecular docking, and kinetic activity assays coupled with CYP2B6 mutagenesis have done much to characterize this originally neglected monooxygenase. However, a complete understanding of the structural details that make new chemical entities substrates of CYP2B6 is still lacking. Surprisingly little in vitro data has been obtained about the structure-function relationship of amino acids identified to be in the CYP2B6 active site. Since much attention has already been devoted to elucidating the function of CYP2B6 allelic variants, here we review the salient findings of in silico and in vitro studies of the CYP2B6 structure-function relationship with a deliberate focus on the active site. In addition to summarizing these complementary approaches to studying structure-function relationships, we note gaps/challenges in existing data such as the need for more CYP2B6 crystal structures, molecular docking results with various ligands, and data coupling CYP2B6 active site mutagenesis with kinetic parameter measurement under standard expression conditions. Harnessing in silico and in vitro techniques in tandem to understand the CYP2B6 structure-function relationship will likely offer further insights into CYP2B6-mediated metabolism. SIGNIFICANCE STATEMENT: The apparent importance of cytochrome P450 2B6 (CYP2B6) in the metabolism of various xenobiotics and endogenous molecules has grown since its discovery with many in silico and in vitro studies offering a partial description of its structure-function relationship. Determining the structure-function relationship of CYP2B6 is difficult but may be aided by thorough biochemical investigations of the CYP2B6 active site that provide a more complete pharmacological understanding of this important enzyme.
Collapse
Affiliation(s)
- Ethan D Angle
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, Azusa Pacific University, Azusa, California (E.D.A., P.M.C.) and Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa (E.D.A.)
| | - Philip M Cox
- Department of Biology and Chemistry, College of Liberal Arts and Sciences, Azusa Pacific University, Azusa, California (E.D.A., P.M.C.) and Roy J. and Lucille A. Carver College of Medicine University of Iowa, Iowa City, Iowa (E.D.A.)
| |
Collapse
|
5
|
Mast N, Li Y, Pikuleva IA. Increased Acetylcholine Levels and Other Brain Effects in 5XFAD Mice after Treatment with 8,14-Dihydroxy Metabolite of Efavirenz. Int J Mol Sci 2022; 23:ijms23147669. [PMID: 35887013 PMCID: PMC9317559 DOI: 10.3390/ijms23147669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 02/08/2023] Open
Abstract
Efavirenz (EFV), an FDA-approved anti-HIV drug, has off-target binding to CYP46A1, the CNS enzyme which converts cholesterol to 24-hydroxycholesterol. At small doses, EFV allosterically activates CYP46A1 in mice and humans and mitigates some of the Alzheimer's disease manifestations in 5XFAD mice, an animal model. Notably, in vitro, all phase 1 EFV hydroxymetabolites activate CYP46A1 as well and bind either to the allosteric site for EFV, neurotransmitters or both. Herein, we treated 5XFAD mice with 8,14-dihydroxyEFV, the binder to the neurotransmitter allosteric site, which elicits the highest CYP46A1 activation in vitro. We found that treated animals of both sexes had activation of CYP46A1 and cholesterol turnover in the brain, decreased content of the amyloid beta 42 peptide, increased levels of acetyl-CoA and acetylcholine, and altered expression of the brain marker proteins. In addition, male mice had improved performance in the Barnes Maze test and increased expression of the acetylcholine-related genes. This work expands our knowledge of the beneficial CYP46A1 activation effects and demonstrates that 8,14-dihydroxyEFV crosses the blood-brain barrier and has therapeutic potential as a CYP46A1 activator.
Collapse
|
6
|
Mast N, Fotinich A, Pikuleva IA. The Hydroxylation Position Rather than Chirality Determines How Efavirenz Metabolites Activate Cytochrome P450 46A1 In Vitro. Drug Metab Dispos 2022; 50:923-930. [PMID: 35489779 PMCID: PMC11022896 DOI: 10.1124/dmd.122.000874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/05/2022] [Indexed: 11/22/2022] Open
Abstract
(S)-Efavirenz (EFV) is a reverse transcriptase inhibitor and an antiviral drug. In addition, (S)-EFV can interact off target with CYP46A1, the major cholesterol hydroxylating enzyme in the mammalian brain, and allosterically activate CYP46A1 at a small dose in mice and humans. Studies with purified CYP46A1 identified two allosteric sites on the enzyme surface, one for (S)-EFV and the second site for L-glutamate (Glu), a neurotransmitter that also activates CYP46A1 either alone or in the presence of (S)-EFV. Previously, we found that racemic (rac)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, (S)-8-hydroxyefavirenz, and (rac)-8,14-dihydroxyefavirenz, compounds with the hydroxylation positions corresponding to the metabolism of (S)-EFV in the liver, activated CYP46A1 in vitro. Yet, these compounds differed from (S)-EFV in how they allosterically interacted with CYP46A1. Herein, we further characterized (rac)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, (S)-8-hydroxyefavirenz, and (rac)-8,14-dihydroxyefavirenz, and, in addition, (R)-EFV, (S)-7-hydroxyefavirenz, (rac)-7,8-dihydroxyefavirenz, (S)-7,8-dihydroxyefavirenz, and (S)-8,14-dihydroxyefavirenz for activation and binding to CYP46A1 in vitro. We found that the spatial configuration of all tested compounds neither affected the CYP46A1 activation nor the sites of binding to CYP46A1. Yet, the hydroxylation position determined whether the hydroxylated metabolite interacted with the allosteric site for (S)-EFV [(R)-EFV, (rac)-7,8-dihydroxyefavirenz, and (S)-7,8-dihydroxyefavirenz], L-Glu [(rac)- and (S)-8,14-dihydroxyefavirenz], or both [(rac)-7-hydroxyefavirenz, (S)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, and (S)-8-hydroxyefavirenz]. This difference in binding to the allosteric sites determined, in turn, how CYP46A1 activity was changed in the coincubations with (S)-EFV and either its metabolite or L-Glu. The results suggest EFV metabolites that could be more potent for CYP46A1 activation in vivo than (S)-EFV. SIGNIFICANCE STATEMENT: This study found that not only efavirenz but also all its hydroxylated metabolites allosterically activate CYP46A1 in vitro. The enzyme activation depended on the hydroxylation position but not the metabolite spatial configuration and involved either one or two allosteric sites-for efavirenz, L-glutamate, or both. The results suggest that the hydroxylated efavirenz metabolites may differ from efavirenz in how they interact with the CYP46A1 allosteric and active sites.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Anna Fotinich
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
7
|
Shi N, Zheng Q, Zhang H. Molecular Basis of the Recognition of Cholesterol by Cytochrome P450 46A1 along the Major Access Tunnel. ACS Chem Neurosci 2022; 13:1526-1533. [PMID: 35438962 DOI: 10.1021/acschemneuro.1c00866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
CYP46A1 is an important potential target for the treatment of Alzheimer's disease (AD), which is the most common neurodegenerative disease among older individuals. However, the binding mechanism between CYP46A1 and substrate cholesterol (CH) has not been clarified and will not be conducive to the research of relevant drug molecules. In this study, we integrated molecular docking, molecular dynamics (MD) simulations, and adaptive steered MD simulations to explore the recognition and binding mechanism of CYP46A1 with CH. Two key factors affecting the interaction between CH and CYP46A1 are determined: one is a hydrophobic cavity formed by seven hydrophobic residues (F80, Y109, L112, I222, W368, F371, and T475), which provides nonpolar interactions to stabilize CH, and the other is a hydrogen bond formed by H81 and CH, which ensures the binding direction of CH. In addition, the tunnel analysis results show that tunnel 2a is identified as the primary pathway of CH. The entry of CH induces tunnel 2e to close and tunnel w to open. Our results may provide effective clues for the design of drugs based on the substrate for AD and improve our understanding of the structure-function of CYP46A1.
Collapse
Affiliation(s)
- Na Shi
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| | - Qingchuan Zheng
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun 130023, China
| | - Hongxing Zhang
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| |
Collapse
|
8
|
Ojagbemi A. HIV Associated Neurocognitive Disorders Subsidence Through Citalopram Addition in Anti-retroviral Therapy (HANDS-CARE): A Concept Note. Front Neurol 2021; 12:658705. [PMID: 34381409 PMCID: PMC8350562 DOI: 10.3389/fneur.2021.658705] [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: 01/26/2021] [Accepted: 06/18/2021] [Indexed: 11/13/2022] Open
Abstract
There is a pressing need to effectively manage HIV Associated Neurocognitive Disorders (HAND) in sub-Saharan Africa (SSA) where the burden is among the highest in the world. Contemporary approaches based on the use of Highly Active Antiretroviral Therapy (HAART) alone are inadequate interventions for HAND, especially in SSA where there is limited availability of the required combinations of HAART for effective central nervous system penetration and where many currently prescribed agents, including efavirenz, have neurotoxicity as a major drawback. This article reviews data supporting the rationale for additive citalopram in antiretroviral therapy as a latent approach to abate HAND. It proposes the conduct of a HIV Associated Neurocognitive Disorders Subsidence through Citalopram addition in Anti-Retroviral therapy (HANDS-CARE) pilot feasibility trial (RCT) to assess whether the adjunctive use of citalopram, a widely prescribed serotonergic antidepressant, will lead to a meaningful improvement in neurocognitive functioning and quality of life in patients with HAND who are receiving HAART. A preliminarily feasible and efficacy-suggesting HANDS-CARE trial could generate statistical, clinical and operational data necessary to design and conduct a future definitive RCT. If successful, this intervention will be applicable to resource-limited settings as well as developed countries. Effective management of HAND will improve the quality of life of HIV patients, and reduce the cost of managing the disease.
Collapse
Affiliation(s)
- Akin Ojagbemi
- Department of Psychiatry, World Health Organization Collaborating Centre for Research and Training in Mental Health, Substance Abuse and Neuroscience, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
9
|
Andriguetti NB, Barratt DT, Tucci J, Pumuye P, Somogyi AA. Instability of Efavirenz Metabolites Identified During Method Development and Validation. J Pharm Sci 2021; 110:3362-3366. [PMID: 34175301 DOI: 10.1016/j.xphs.2021.06.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 11/27/2022]
Abstract
Accurate quantification of efavirenz metabolites in patient samples is required to investigate their potential contribution to efavirenz adverse events. This study aimed to validate a LC-MS/MS method to quantify and investigate the stability of efavirenz and metabolites in human plasma. Compounds were extracted from plasma by supported liquid extraction and resolved on a C18 column. Validation was performed following FDA bioanalytical method validation guidelines. Stability under common conditions of sample pre-treatment and storage were assessed. Efavirenz and 8-hydroxyefavirenz were stable for all conditions tested. 7-Hydroxyefavirenz and 8,14-dihydroxyefavirenz were not stable in plasma at room temperature for 24 h (46%-69% loss), -20°C for 90 days (17%-50% loss), or 60°C for 1 h (90%-95% loss). Efavirenz and 8-hydroxyefavirenz concentrations in HIV/AIDS patient (n=5) plasma prepared from pre-treated (60°C for 1 h) whole blood varied from 517-8564 ng/mL and 131-813 ng/mL, respectively. 7-Hydroxyefavirenz and 8,14-dihydroxyefavirenz concentrations were below validated lower limits of quantification (0.25 and 0.5 ng/mL, respectively), most likely due to sample pre-treatment. This is the first report of 7-hydroxyefavirenz and 8,14-dihydroxyefavirenz instability under conditions commonly used in preparation of samples from HIV/AIDS patients. Alternative biosafety measures to heat pre-treatment must therefore be used for accurate quantification of plasma 7-hydroxyefavirenz and 8,14-dihydroxyefavirenz.
Collapse
Affiliation(s)
| | - Daniel T Barratt
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Joseph Tucci
- Department of Pharmacy & Biomedical Sciences, La Trobe University Bendigo Campus, Bendigo, Australia
| | - Paul Pumuye
- School of Medicine and Health Sciences, University of Papua New Guinea, National Capital District, Papua New Guinea
| | - Andrew A Somogyi
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| |
Collapse
|
10
|
Yu ZJ, Mosher EP, Bumpus NN. Pharmacogenomics of Antiretroviral Drug Metabolism and Transport. Annu Rev Pharmacol Toxicol 2020; 61:565-585. [PMID: 32960701 DOI: 10.1146/annurev-pharmtox-021320-111248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antiretroviral therapy has markedly reduced morbidity and mortality for persons living with human immunodeficiency virus (HIV). Individual tailoring of antiretroviral regimens has the potential to further improve the long-term management of HIV through the mitigation of treatment failure and drug-induced toxicities. While the mechanisms underlying anti-HIV drug adverse outcomes are multifactorial, the application of drug-specific pharmacogenomic knowledge is required in order to move toward the personalization of HIV therapy. Thus, detailed understanding of the metabolism and transport of antiretrovirals and the influence of genetics on these pathways is important. To this end, this review provides an up-to-date overview of the metabolism of anti-HIV therapeutics and the impact of genetic variation in drug metabolism and transport on the treatment of HIV. Future perspectives on and current challenges in pursuing personalized HIV treatment are also discussed.
Collapse
Affiliation(s)
- Zaikuan J Yu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| | - Eric P Mosher
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| | - Namandjé N Bumpus
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| |
Collapse
|
11
|
Semen Extracellular Vesicles From HIV-1-Infected Individuals Inhibit HIV-1 Replication In Vitro, and Extracellular Vesicles Carry Antiretroviral Drugs In Vivo. J Acquir Immune Defic Syndr 2020; 83:90-98. [PMID: 31809364 DOI: 10.1097/qai.0000000000002233] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs) are cell-derived vesicles with diverse functions in intercellular communication including disease and infection, and EVs seem to influence HIV-1 pathogenesis. EVs isolated from HIV-1-uninfected semen (SE), but not blood (BE), contain factors that interfere with HIV-1 infection and replication in target cells. The reason for this dichotomy is unknown. Furthermore, the effect of HIV-1 infection and antiretroviral (ARV) drugs on the anti-HIV-1 effects of SE and BE is unknown. Here, we characterize EVs and EV-free plasma isolated from HIV-infected donor semen and blood and their effects on HIV infection. METHODS EVs and EV-free plasma were purified from autologous blood and semen of HIV-negative, HIV-infected antiretroviral therapy (ART)-naïve, and HIV-infected ART-treated participants. HIV infection was assessed in a TZM-bl cell reporter system. ARV concentrations were analyzed using liquid chromatography-mass spectrometry. RESULTS SE isolated from both HIV-negative and HIV-infected, ART-naïve donors inhibited HIV-1 infection, but BE and semen and blood EV-free plasma did not. By contrast, BE, SE, and EV-free plasma from HIV-infected, ART-treated donors inhibited HIV-1. Importantly, exosomes isolated from ART-treated donors contained concentrations of ARV drugs (ART-EVs) at biologically relevant inhibitory levels. CONCLUSIONS The HIV-1-inhibitory phenotype of SE is independent of donor HIV-1 or ART status, and ARV drugs and their metabolites are SE- and BE-associated in vivo.
Collapse
|
12
|
Mast N, Verwilst P, Wilkey CJ, Guengerich FP, Pikuleva IA. In Vitro Activation of Cytochrome P450 46A1 (CYP46A1) by Efavirenz-Related Compounds. J Med Chem 2019; 63:6477-6488. [PMID: 31617715 PMCID: PMC7226586 DOI: 10.1021/acs.jmedchem.9b01383] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
Cytochrome P450 46A1 (CYP46A1) is a central nervous system-specific
enzyme, which catalyzes cholesterol 24-hydroxylation. Currently CYP46A1
is being evaluated in a clinical trial for activation by small doses
of the anti-HIV drug efavirenz. Eight efavirenz-related compounds
were investigated for CYP46A1 activation in vitro, induction of a
CYP46A1 spectral response, spectral Kd values, interaction with the P450 allosteric sites, and a model
of binding to the enzyme active site. We gained insight into structure–activity
relationships of efavirenz for CYP46A1 activation and found that the
investigated efavirenz primary metabolites are stronger and better
activators of CYP46A1 than efavirenz. We also established that CYP46A1
is activated by racemates and that a conformational-selection mechanism
is operative in CYP46A1. The results suggest structural modifications
of efavirenz to further increase CYP46A1 activation without inhibition
at high compound concentrations. It is possible that not only efavirenz
but its metabolites activate CYP46A1 in vivo.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Peter Verwilst
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Clayton J Wilkey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
13
|
Wang PF, Neiner A, Kharasch ED. Efavirenz Metabolism: Influence of Polymorphic CYP2B6 Variants and Stereochemistry. Drug Metab Dispos 2019; 47:1195-1205. [PMID: 31324697 PMCID: PMC6756292 DOI: 10.1124/dmd.119.086348] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 07/10/2019] [Indexed: 01/11/2023] Open
Abstract
Efavirenz (more specifically the S-enantiomer) is a cornerstone antiretroviral therapy for treatment of HIV infection. The major primary metabolite is S-8-hydroxyefavirenz, which does not have antiretroviral activity but is neurotoxic. Cytochrome P450 2B6 (CYP2B6) is the major enzyme catalyzing S-8-hydroxyefavirenz formation. CYP2B6 genetics and drug interactions are major determinants of clinical efavirenz disposition and dose adjustment. In addition, as a prototypic CYP2B6 substrate, S-efavirenz and analogs can inform on the structure, activity, catalytic mechanisms, and stereoselectivity of CYP2B6. Metabolism of R-efavirenz by CYP2B6 remains unexplored. This investigation assessed S-efavirenz metabolism by clinically relevant CYP2B6 genetic variants. This investigation also evaluated R-efavirenz hydroxylation by wild-type CYP2B6.1 and CYP2B6 variants. S-Efavirenz 8-hydroxylation by wild-type CYP2B6.1 and variants exhibited positive cooperativity and apparent cooperative substrate inhibition. On the basis of Clmax values, relative activities for S-efavirenz 8-hydroxylation were in the order CYP2B6.4 > CYP2B6.1 ≈ CYP2B6.5 ≈ CYP2B6.17 > CYP2B6.6 ≈ CYP2B6.7 ≈ CYP2B6.9 ≈ CYP2B6.19 ≈ CYP2B6.26; CYP2B6.16 and CYP2B6.18 showed minimal activity. Rates of R-efavirenz metabolism were approximately 1/10 those of S-efavirenz for wild-type CYP2B6.1 and variants. On the basis of Clmax values, there was 14-fold enantioselectivity (S > R-efavirenz) for wild-type CYP2B6.1, and 5- to 22-fold differences for other CYP2B6 variants. These results show that both CYP2B6 516G > T (CYP2B6*6 and CYP2B6*9) and 983T > C (CYP2B6*16 and CYP2B6*18) polymorphisms cause canonical diminishment or loss-of-function variants for S-efavirenz 8-hydroxylation, provide a mechanistic basis for known clinical pharmacogenetic differences in efavirenz disposition, and may predict additional clinically important variant alleles. Efavirenz is the most stereoselective CYP2B6 drug substrate yet identified and may be a useful probe for the CYP2B6 active site and catalytic mechanisms. SIGNIFICANCE STATEMENT: Clinical disposition of the antiretroviral S-efavirenz is affected by CYP2B6 polymorphisms. Expressed CYP2B6 with 516G>T (CYP2B6*6 and CYP2B6*9), and 983T>C (CYP2B6*16 and CYP2B6*18) polymorphisms had a diminishment or loss of function for efavirenz 8-hydroxylation. This provides a mechanistic basis for efavirenz clinical pharmacogenetics and may predict additional clinically important variant alleles. Efavirenz metabolism showed both cooperativity and cooperative substrate inhibition. With greater than 10-fold enantioselectivity (S- vs. R- metabolism), efavirenz is the most stereoselective CYP2B6 drug substrate yet identified. These findings may provide mechanistic insights.
Collapse
Affiliation(s)
- Pan-Fen Wang
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| | - Alicia Neiner
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri.
| | - Evan D Kharasch
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
14
|
Decloedt EH, Sinxadi PZ, van Zyl GU, Wiesner L, Khoo S, Joska JA, Haas DW, Maartens G. Pharmacogenetics and pharmacokinetics of CNS penetration of efavirenz and its metabolites. J Antimicrob Chemother 2019; 74:699-709. [PMID: 30535366 PMCID: PMC6376850 DOI: 10.1093/jac/dky481] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/11/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND There are limited data on the pharmacogenetics and pharmacokinetics of the CNS penetration of efavirenz. OBJECTIVES We investigated genetic polymorphisms associated with CSF concentrations of efavirenz and its metabolites and explored the relationships with neurocognitive performance. METHODS We included 47 HIV-infected South African black adults with and without HIV-associated neurocognitive disorder on efavirenz/tenofovir/emtricitabine and collected paired plasma-CSF samples. We considered 2049 SNPs, including SNPs known to affect plasma efavirenz exposure, from potentially relevant genes (ABCC5, ABCG2, ABCB1, SLCO2B1, SCLO1A2, ABCC4, CYP2B6 and CYP2A6) and 880 met a linkage disequilibrium (LD)-pruning threshold. RESULTS We identified 9 slow, 21 intermediate and 17 extensive metabolizers. The CYP2B6 983 genotype in multivariate analyses predicted log10-transformed concentrations of plasma efavirenz (β = 0.38, P = 2.7 × 10-03), plasma 7-hydroxy-efavirenz (β = 0.59, P = 3.7 × 10-03), plasma 8-hydroxy-efavirenz:efavirenz ratio (β = -0.31, P = 1.8 × 10-04) and CSF efavirenz (β = 0.36, P = 0.01). Lower plasma 7-hydroxy-efavirenz concentrations were independently associated with CYP2A6 rs10853742 (β = -0.55, P = 3.5 × 10-05), ABCB1 rs115780656 (β = -0.65, P = 4.1 × 10-05) and CYP2A6 -48A→C (β = -0.59, P = 0.01). CYP2A6 -48A→C was independently associated with higher CSF 8-hydroxy-efavirenz:efavirenz ratio (β = 0.54, P = 0.048). CYP2B6 rs2279345 polymorphism was associated with lower plasma 7-hydroxy-efavirenz:efavirenz ratio in multivariate analyses (P < 0.05). No polymorphisms were associated with CSF:plasma ratios of efavirenz, plasma or CSF concentrations of 8-hydroxy-efavirenz or neurocognitive performance. CONCLUSIONS We identified novel genetic associations with plasma efavirenz, plasma 7-hydroxy-efavirenz, plasma 7-hydroxy-efavirenz:efavirenz ratio, plasma 8-hydroxy-efavirenz:efavirenz ratio, CSF efavirenz and CSF 8-hydroxy-efavirenz:efavirenz ratio.
Collapse
Affiliation(s)
- Eric H Decloedt
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Phumla Z Sinxadi
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Gert U van Zyl
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University and National Health Laboratory Service, Cape Town, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Saye Khoo
- Institute of Translational Medicine, University of Liverpool and Royal Liverpool University Hospital, Liverpool, UK
| | - John A Joska
- Division of Neuropsychiatry, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - David W Haas
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Internal Medicine, Meharry Medical College, Nashville, TN, USA
| | - Gary Maartens
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
15
|
Van den Hof M, Blokhuis C, Cohen S, Scherpbier HJ, Wit FWNM, Pistorius MCM, Kootstra NA, Teunissen CE, Mathot RAA, Pajkrt D. CNS penetration of ART in HIV-infected children. J Antimicrob Chemother 2019; 73:484-489. [PMID: 29126299 DOI: 10.1093/jac/dkx396] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 09/29/2017] [Indexed: 12/29/2022] Open
Abstract
Background Paediatric data on CNS penetration of antiretroviral drugs are scarce. Objectives To evaluate CNS penetration of antiretroviral drugs in HIV-infected children and explore associations with neurocognitive function. Patients and methods Antiretroviral drug levels were measured in paired CSF and blood samples of clinically stable HIV-infected children between 8 and 18 years old on long-term combined ART. Plasma drug concentrations were corrected for protein binding. We evaluated CNS penetration using CSF/plasma ratios and compared CSF concentrations with the IC50 as a surrogate marker for effectiveness. Blood-brain barrier permeability was assessed for possible confounding. Associations with neurocognitive function were explored using linear regression analysis. Results Median CSF/plasma ratios (IQR) were: lopinavir 0.059 (0.024-0.157, n = 7), efavirenz 0.681 (0.555-0.819, n = 12), tenofovir 0.021 (0.020-0.024, n = 4), lamivudine 0.464 (0.331-0.607, n = 17), emtricitabine 0.365 (0.343-0.435, n = 3), nevirapine 1.203 (n = 1), zidovudine 0.718 (0.711-1.227, n = 5) and abacavir 1.344 (0.670-2.450, n = 10). CSF concentrations were below the IC50 for tenofovir (100%), emtricitabine (100%), abacavir (50%) and zidovudine (17%). Lamivudine, lopinavir, efavirenz and nevirapine concentrations were all above the IC50. All participants were virologically suppressed in blood and CSF. CSF drug concentrations were not associated with blood-brain barrier permeability or neurocognitive function. Conclusions We showed adequate CSF concentrations of lamivudine, lopinavir, efavirenz and nevirapine, and potential suboptimal CSF concentrations of tenofovir, abacavir and emtricitabine in long-term treated HIV-infected children. None the less, the use of combined antiretroviral drugs led to adequate viral suppression.
Collapse
Affiliation(s)
- Malon Van den Hof
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Charlotte Blokhuis
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sophie Cohen
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Henriette J Scherpbier
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Ferdinand W N M Wit
- Department of Internal Medicine, Division of Infectious Diseases, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.,Department of Global Health, Academic Medical Center, University of Amsterdam, and Amsterdam Institute for Global Health and Development, Pietersbergweg 7, 1105 BM The Netherlands.,HIV Monitoring Foundation, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - M C M Pistorius
- Department of Hospital Pharmacy-Clinical Pharmacology Unit, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Academic Medical Centre, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, VU University Medical Centre and Neurocampus Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Ron A A Mathot
- Department of Hospital Pharmacy-Clinical Pharmacology Unit, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
16
|
Heck CJS, Hamlin AN, Bumpus NN. Efavirenz and Efavirenz-like Compounds Activate Human, Murine, and Macaque Hepatic IRE1 α-XBP1. Mol Pharmacol 2019; 95:183-195. [PMID: 30442673 PMCID: PMC6324649 DOI: 10.1124/mol.118.113647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
Efavirenz (EFV), a widely used antiretroviral drug, is associated with idiosyncratic hepatotoxicity and dyslipidemia. Here we demonstrate that EFV stimulates the activation in primary hepatocytes of key cell stress regulators: inositol-requiring 1α (IRE1α) and X-box binding protein 1 (XBP1). Following EFV exposure, XBP1 splicing (indicating activation) was increased 35.7-fold in primary human hepatocytes. In parallel, XBP1 splicing and IRE1α phosphorylation (p-IRE1α, active IRE1α) were elevated 36.4-fold and 4.9-fold, respectively, in primary mouse hepatocytes. Of note, with EFV treatment, 47.2% of mouse hepatocytes were apoptotic; which was decreased to 23.9% in the presence of STF 083010, an inhibitor of XBP1 splicing. Experiments performed using pregnane X receptor (PXR)-null mouse hepatocytes revealed that EFV-mediated XBP1 splicing and hepatocyte death were not dependent on PXR, which is a nuclear receptor transcription factor that plays a crucial role in the cellular response to xenobiotics. Interestingly, incubation with the primary metabolite of EFV, 8-hydroxyefavirenz (8-OHEFV), only resulted in 10.3- and 2.9-fold increased XBP1 splicing in human and mouse hepatocytes and no change in levels of p-IRE1α in mouse hepatocytes. To further probe the structure-activity relationship of IRE1α-XBP1 activation by EFV, 16 EFV analogs were employed. Of these, an analog in which the EFV alkyne is replaced with an alkene and an analog in which the oxazinone oxygen is replaced by a carbon stimulated XBP1 splicing in human, mouse, and macaque hepatocytes. These data demonstrate that EFV and compounds sharing the EFV scaffold can activate IRE1α-XBP1 across human, mouse, and macaque species.
Collapse
Affiliation(s)
- Carley J S Heck
- Department of Pharmacology and Molecular Sciences (C.J.S.H., N.N.B.) and Department of Medicine, Division of Clinical Pharmacology (A.N.H., N.N.B.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Allyson N Hamlin
- Department of Pharmacology and Molecular Sciences (C.J.S.H., N.N.B.) and Department of Medicine, Division of Clinical Pharmacology (A.N.H., N.N.B.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Namandjé N Bumpus
- Department of Pharmacology and Molecular Sciences (C.J.S.H., N.N.B.) and Department of Medicine, Division of Clinical Pharmacology (A.N.H., N.N.B.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
17
|
Narayanan B, Lade JM, Heck CJ, Dietz KD, Wade H, Bumpus NN. Probing Ligand Structure-Activity Relationships in Pregnane X Receptor (PXR): Efavirenz and 8-Hydroxyefavirenz Exhibit Divergence in Activation. ChemMedChem 2018; 13:736-747. [PMID: 29430850 PMCID: PMC6081956 DOI: 10.1002/cmdc.201700730] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/06/2018] [Indexed: 12/24/2022]
Abstract
Efavirenz (EFV), an antiretroviral that interacts clinically with co-administered drugs via activation of the pregnane X receptor (PXR), is extensively metabolized by the cytochromes P450. We tested whether its primary metabolite, 8-hydroxyEFV (8-OHEFV) can activate PXR and potentially contribute to PXR-mediated drug-drug interactions attributed to EFV. Luciferase reporter assays revealed that despite only differing from EFV by an oxygen atom, 8-OHEFV does not activate PXR. Corroborating this, treatment with EFV for 72 h elevated the mRNA abundance of the PXR target gene, Cyp3a11, by approximately 28-fold in primary hepatocytes isolated from PXR-humanized mice, whereas treatment with 8-OHEFV did not result in a change in Cyp3A11 mRNA levels. FRET-based competitive binding assays and isothermal calorimetry demonstrated that even with the lack of ability to activate PXR, 8-OHEFV displays an affinity for PXR (IC50 12.1 μm; KD 7.9 μm) nearly identical to that of EFV (IC50 18.7 μm; KD 12.5 μm). The use of 16 EFV analogues suggest that other discreet changes to the EFV structure beyond the 8-position are well tolerated. Molecular docking simulations implicate an 8-OHEFV binding mode that may underlie its divergence in PXR activation from EFV.
Collapse
Affiliation(s)
- Bhargavi Narayanan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe Street Hunterian 709 Baltimore, MD, USA
| | - Julie M. Lade
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St Biophysics 307 Baltimore, MD, USA
| | - Carley J.S. Heck
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N Wolfe St Biophysics 307 Baltimore, MD, USA
| | - Kevin D. Dietz
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe Street Hunterian 709 Baltimore, MD, USA
| | - Herschel Wade
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N Wolfe Street Hunterian 709 Baltimore, MD, USA
| | - Namandjé N. Bumpus
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, 725 N Wolfe St Biophysics 307 Baltimore, MD, USA
| |
Collapse
|
18
|
Zhuo X, Wang YZ, Yeung KS, Zhu J, Huang XS, Parcella KE, Eastman KJ, Kadow JF, Meanwell NA, Shu YZ, Johnson BM. Bioactivation of cyclopropyl rings by P450: an observation encountered during the optimisation of a series of hepatitis C virus NS5B inhibitors. Xenobiotica 2017; 48:1215-1226. [PMID: 29182424 DOI: 10.1080/00498254.2017.1409915] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
1. Due to its unique C-C and C-H bonding properties, conformational preferences and relative hydrophilicity, the cyclopropyl ring has been used as a synthetic building block in drug discovery to modulate potency and drug-like properties. During an effort to discover inhibitors of the hepatitis C virus non-structural protein 5B with improved potency and genotype-coverage profiles, the use of a pyrimidinylcyclopropylbenzamide moiety linked to a C6-substituted benzofuran or azabenzofuran core scaffold was explored in an effort to balance antiviral potency and metabolic stability. 2. In vitro metabolism studies of two compounds from this C6-substituted series revealed an NADPH-dependent bioactivation pathway leading to the formation of multiple glutathione (GSH) conjugates. Analysis of these conjugates by LC-MS and NMR demonstrated that the cyclopropyl group was the site of bioactivation. Based on the putative structures and molecular weights of the cyclopropyl-GSH conjugates, a multi-step mechanism was proposed to explain the formation of these metabolites by P450. This mechanism involves hydrogen atom abstraction to form a cyclopropyl radical, followed by a ring opening rearrangement and reaction with GSH. 3. These findings provided important information to the medicinal chemistry team which responded by replacing the cyclopropyl ring with a gem-dimethyl group. Subsequent compounds bearing this feature were shown to avert the bioactivation pathways in question.
Collapse
Affiliation(s)
- Xiaoliang Zhuo
- a Departments of Pharmaceutical Candidate Optimisation , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA and
| | - Ying-Zi Wang
- a Departments of Pharmaceutical Candidate Optimisation , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA and
| | - Kap-Sun Yeung
- b Discovery Chemistry and Molecular Technologies , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA
| | - Juliang Zhu
- b Discovery Chemistry and Molecular Technologies , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA
| | - Xiaohua Stella Huang
- a Departments of Pharmaceutical Candidate Optimisation , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA and
| | - Kyle E Parcella
- b Discovery Chemistry and Molecular Technologies , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA
| | - Kyle J Eastman
- b Discovery Chemistry and Molecular Technologies , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA
| | - John F Kadow
- b Discovery Chemistry and Molecular Technologies , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA
| | - Nicholas A Meanwell
- b Discovery Chemistry and Molecular Technologies , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA
| | - Yue-Zhong Shu
- a Departments of Pharmaceutical Candidate Optimisation , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA and
| | - Benjamin M Johnson
- a Departments of Pharmaceutical Candidate Optimisation , Bristol-Myers Squibb Research and Development , Wallingford , CT , USA and
| |
Collapse
|
19
|
Population Pharmacokinetic Model Linking Plasma and Peripheral Blood Mononuclear Cell Concentrations of Efavirenz and Its Metabolite, 8-Hydroxy-Efavirenz, in HIV Patients. Antimicrob Agents Chemother 2017; 61:AAC.00207-17. [PMID: 28559276 DOI: 10.1128/aac.00207-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/21/2017] [Indexed: 01/27/2023] Open
Abstract
The objectives of this study were to characterize the population pharmacokinetics (PK) of efavirenz (EFV) and 8-hydroxy-efavirenz (8OHEFV) in plasma and peripheral blood mononuclear cells (PBMCs) and to explore covariates affecting the PK parameters. Fifty-one patients had steady-state 0-to-24-h concentrations of EFV and 8OHEFV in plasma with corresponding concentrations in PBMCs, while 261 patients had one or two sparse concentrations at 16 ± 1 h postdose at weeks 4 and/or 16. The pharmacogenetic markers CYP2B6*6, CYP3A5*3, CYP3A5*6, UGT2B7*2, ABCB1 (3435C→T, 3842A→G), OATP1B1*1B, and OATP1B1*5, the presence of a rifampin-based antituberculosis (anti-TB) regimen, baseline body weight and organ function values, and demographic factors were explored as covariates. EFV concentration data were well described by a two-compartment model with first-order absorption (Ka ) and absorption lag time (Alag) (Ka = 0.2 h-1; Alag = 0.83 h; central compartment clearance [CLc/F] for CYP2B6*1/*1 = 18 liters/h, for CYP2B6*1/*6 = 14 liters/h, and for CYP2B6*6/*6 = 8.6 liters/h) and PBMCs as a peripheral compartment. EFV transfer from plasma to PBMCs was first order (CLp/F = 32 liters/h), followed by capacity-limited return (Vmax = 4,400 ng/ml/h; Km = 710 ng/ml). Similarly, 8OHEFV displayed a first-order elimination and distribution to PBMCs, with a capacity-limited return to plasma. No covariate relationships resulted in a significant explanation of interindividual variability (IIV) on the estimated PK parameters of EFV and 8OHEFV, except for CYP2B6*6 genotypes, which were consistent with prior evidence. Both EFV and 8OHEFV accumulated to higher concentrations in PBMCs than in plasma and were well described by first-order input and Michaelis-Menten kinetics removal from PBMCs. CYP2B6*6 genotype polymorphisms were associated with decreased EFV and 8OHEFV clearance.
Collapse
|
20
|
Unmasking efavirenz neurotoxicity: Time matters to the underlying mechanisms. Eur J Pharm Sci 2017; 105:47-54. [PMID: 28487145 DOI: 10.1016/j.ejps.2017.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/06/2017] [Accepted: 05/05/2017] [Indexed: 12/14/2022]
Abstract
Efavirenz is an anti-HIV drug that presents relevant short- and long-term central nervous system adverse reactions. Its main metabolite (8-hydroxy-efavirenz) was demonstrated to be a more potent neurotoxin than efavirenz itself. This work was aimed to understand how efavirenz biotransformation to 8-hydroxy-efavirenz is related to its short- and long-term neuro-adverse reactions. To access those mechanisms, the expression and activity of Cyp2b enzymes as well as the thiolomic signature (low molecular weight thiols plus S-thiolated proteins) were longitudinally evaluated in the hepatic and brain tissues of rats exposed to efavirenz during 10 and 36days. Efavirenz and 8-hydroxy-efavirenz plasma concentrations were monitored at the same time points. Cyp2b induction had a delayed onset in liver (p<0.001), translating into increases in Cyp2b activity in liver and 8-hydroxy-efavirenz plasma concentration (p<0.001). Moreover, an increase in S-cysteinyl-glycinylated proteins (p<0.001) and in free low molecular weight thiols was also observed in liver. A distinct scenario was observed in hippocampus, which showed an underexpression of Cyp2b as well as a decrease in S-cysteinylated and S-glutathionylated proteins. Additionally, the observed changes in tissues were associated with a marked increase of S-glutathionylation in plasma. Our data suggest that the time course of efavirenz biotransformation results from different mechanisms for its short- and long-term neurotoxicity. The difference in the redox profile between liver and hippocampus might explain why, despite being mostly metabolized by the liver, this drug is neurotoxic. If translated to clinical practice, this evidence will have important implications in efavirenz short- and long-term neurotoxicity prevention and management.
Collapse
|
21
|
Sandkovsky U, Podany AT, Fletcher CV, Owen A, Felton-Coleman A, Winchester LC, Robertson K, Swindells S. Impact of efavirenz pharmacokinetics and pharmacogenomics on neuropsychological performance in older HIV-infected patients. J Antimicrob Chemother 2016; 72:200-204. [PMID: 27655857 DOI: 10.1093/jac/dkw403] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/16/2016] [Accepted: 08/26/2016] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Pharmacokinetics (PK) and pharmacodynamics of efavirenz and its 8-hydroxy metabolite (8-OH-efavirenz) have not been robustly evaluated in older HIV-infected persons. OBJECTIVES We investigated relationships between neuropsychological (NP) performance and efavirenz and 8-OH-efavirenz PK in HIV-infected individuals >50 years of age. METHODS A cross-sectional study of HIV-infected adults on an efavirenz-containing regimen. The 12 and 18 h post-dose plasma efavirenz and 8-OH-efavirenz were quantified. CYP2B6 polymorphisms were investigated. Participants underwent neuropsychological tests; surveys were used for depression, sleep quality and anxiety. We investigated potential correlations of efavirenz and 8-OH-efavirenz plasma concentrations with NP performance, sleep, depression, anxiety and CYP2B6 polymorphisms. RESULTS Thirty participants (24 men and 6 women) with mean age 57 years (range 50-68). Plasma efavirenz concentrations did not correlate with NP performance; however, higher plasma 8-OH-efavirenz correlated with better learning (P = 0.002), language (P = 0.002) and total NP z-scores (P = 0.003). No correlation was seen for efavirenz or 8-OH-efavirenz with sleep, anxiety or depression. Median 12 and 18 h efavirenz plasma concentrations were 1967 ng/mL (IQR 1476-2394) and 1676 ng/mL (IQR 1120-2062), respectively. Median 12 and 18 h 8-OH-efavirenz plasma concentrations were 378 ng/mL (IQR 223-589) and 384 ng/mL (IQR 216-621), respectively. CYP2B6 G516T was associated with significantly higher plasma efavirenz at 12 and 18 h (P = 0.02) but not worse NP function. CONCLUSIONS Better neurocognitive functioning was associated with higher 8-OH-efavirenz but not efavirenz plasma concentrations. No correlation was observed with sleep or depression. These findings point to a need for greater understanding of the metabolic profile of efavirenz and 8-OH-efavirenz in plasma and the CNS and relationships with antiviral effect and neurotoxicity.
Collapse
Affiliation(s)
- Uriel Sandkovsky
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony T Podany
- Department of Pharmacy Practice, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Courtney V Fletcher
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pharmacy Practice, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew Owen
- Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Angela Felton-Coleman
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lee C Winchester
- Department of Pharmacy Practice, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kevin Robertson
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Susan Swindells
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
22
|
Consequences of a Chronic Exposure of Cultured Brain Astrocytes to the Anti-Retroviral Drug Efavirenz and its Primary Metabolite 8-Hydroxy Efavirenz. Neurochem Res 2016; 41:3278-3288. [PMID: 27655255 DOI: 10.1007/s11064-016-2059-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 11/27/2022]
Abstract
Efavirenz is a widely prescribed non-nucleoside reverse transcriptase inhibitor for the treatment of HIV infections. To test for potential long-term consequences of efavirenz on brain cells, cultured primary astrocytes were incubated with this substance or with its primary metabolite 8-hydroxy efavirenz for up to 7 days. Both, efavirenz and 8-hydroxy efavirenz caused time- and concentration-dependent cell toxicity and stimulated in subtoxic concentrations the glycolytic flux (glucose consumption and lactate release) in astrocytes. As 8-hydroxy efavirenz was less toxic than efavirenz and stimulated glycolysis in lower concentrations we tested for a potential hydroxylation of efavirenz to 8-hydroxy efavirenz in astrocytes. Analysis of media and cell lysates by HPLC-UV and mass spectrometry revealed that after 3 days of incubation viable astrocytes had accumulated about 17 and 7 % of the applied efavirenz and 8-hydroxy efavirenz, respectively. However, in cultures treated with efavirenz neither 8-hydroxy efavirenz nor any other known metabolite of efavirenz was detectable. These data demonstrate that cultured rat astrocytes efficiently accumulate, but not metabolize, efavirenz and 8-hydroxy efavirenz and that the observed chronic stimulation of glycolysis is mediated by both efavirenz and 8-hydroxy efavirenz.
Collapse
|
23
|
Habtewold A, Aklillu E, Makonnen E, Amogne W, Yimer G, Aderaye G, Bertilsson L, Owen JS, Burhenne J. Long-Term Effect of Rifampicin-Based Anti-TB Regimen Coadministration on the Pharmacokinetic Parameters of Efavirenz and 8-Hydroxy-Efavirenz in Ethiopian Patients. J Clin Pharmacol 2016; 56:1538-1549. [PMID: 27125860 DOI: 10.1002/jcph.756] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/29/2022]
Abstract
We compared the pharmacokinetic (PK) exposure parameters of efavirenz (EFV) and its major inactive metabolite, 8-hydroxy-efavirenz (8-OH-EFV), in an open-label, single-sequence, and parallel design of HIV-infected and tuberculosis (TB)-HIV-coinfected Ethiopian patients in the HIV-TB Pharmagene study with 20 and 33 patients, respectively. Both treatment groups underwent PK sampling following oral 600 mg EFV in week 16 of initiating EFV-based combination antiretroviral therapy. The TB-HIV-coinfected group repeated the PK sampling 8 weeks after stopping rifampin (RIF)-based anti-TB treatment. Between-treatment group analysis indicated no significant effect of RIF-based anti-TB cotreatment on PK exposure parameters of EFV, nor was there a significant effect after controlling for sex or CYP2B6 genotype. However, RIF-based therapy in TB-HIV-coinfected patients had significantly increased 8-OH-EFV PK exposure measures and metabolic ratio relative to HIV-only patients, AUC0-24 greater by 79%. The effect was more prominent in women and CYP2B6*6 carriers in within-sex and CYP2B6 genotype comparisons. Within-subject comparisons for AUC0-24 and Cmax when "on" and "off" RIF-based anti-TB cotreatment showed geometric mean ratios (90% confidence intervals) of 100.5% (98.7%-102.3%) and 100.2% (98.1%-102.4%), respectively, for EFV and 98.6% (95.5%-101.7%-) and 97.6% (92.2%-103.0%), respectively, for 8-OH-EFV. We report no significant influence of RIF-based anti-TB cotherapy on the EFV PK exposure measures. The study also calls for caution related to higher exposure to 8-OH-EFV during simultaneous coadministration of EFV and RIF-based anti-TB regimens, which may be associated with neurotoxicity, particularly in female patients and CYP2B6*6 carriers.
Collapse
Affiliation(s)
- Abiy Habtewold
- Department of Pharmaceutical Sciences, School of Pharmacy, Union University, Jackson, TN, USA.,Division of Clinical Pharmacology, Department of Lab Medicine, Karolinska Institutet Hospital Huddinge, Stockholm, Sweden.,Department of Pharmacology, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Eleni Aklillu
- Section of Pharmacogenetics, Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eyasu Makonnen
- Department of Pharmacology, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Wondwossen Amogne
- Department of Internal Medicine, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Getnet Yimer
- Department of Pharmacology, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Getachew Aderaye
- Department of Internal Medicine, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Leif Bertilsson
- Division of Clinical Pharmacology, Department of Lab Medicine, Karolinska Institutet Hospital Huddinge, Stockholm, Sweden
| | - Joel S Owen
- Department of Pharmaceutical Sciences, School of Pharmacy, Union University, Jackson, TN, USA
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
24
|
Huskey SEW, Zhu CQ, Fredenhagen A, Kühnöl J, Luneau A, Jian Z, Yang Z, Miao Z, Yang F, Jain JP, Sunkara G, Mangold JB, Stein DS. KAE609 (Cipargamin), a New Spiroindolone Agent for the Treatment of Malaria: Evaluation of the Absorption, Distribution, Metabolism, and Excretion of a Single Oral 300-mg Dose of [14C]KAE609 in Healthy Male Subjects. Drug Metab Dispos 2016; 44:672-82. [PMID: 26921387 DOI: 10.1124/dmd.115.069187] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/19/2016] [Indexed: 02/13/2025] Open
Abstract
KAE609 [(1'R,3'S)-5,7'-dichloro-6'-fluoro-3'-methyl-2',3',4',9'-tetrahydrospiro[indoline-3,1'-pyridol[3,4-b]indol]-2-one] is a potent, fast-acting, schizonticidal agent in clinical development for the treatment of malaria. This study investigated the absorption, distribution, metabolism, and excretion of KAE609 after oral administration of [(14)C]KAE609 in healthy subjects. After oral administration to human subjects, KAE609 was the major radioactive component (approximately 76% of the total radioactivity in plasma); M23 was the major circulating oxidative metabolite (approximately 12% of the total radioactivity in plasma). Several minor oxidative metabolites (M14, M16, M18, and M23.5B) were also identified, each accounting for approximately 3%-8% of the total radioactivity in plasma. KAE609 was well absorbed and extensively metabolized, such that KAE609 accounted for approximately 32% of the dose in feces. The elimination of KAE609 and metabolites was primarily mediated via biliary pathways. M23 was the major metabolite in feces. Subjects reported semen discoloration after dosing in prior studies; therefore, semen samples were collected once from each subject to further evaluate this clinical observation. Radioactivity excreted in semen was negligible, but the major component in semen was M23, supporting the rationale that this yellow-colored metabolite was the main source of semen discoloration. In this study, a new metabolite, M16, was identified in all biologic matrices albeit at low levels. All 19 recombinant human cytochrome P450 enzymes were capable of catalyzing the hydroxylation of M23 to form M16 even though the extent of turnover was very low. Thus, electrochemistry was used to generate a sufficient quantity of M16 for structural elucidation. Metabolic pathways of KAE609 in humans are summarized herein and M23 is the major metabolite in plasma and excreta.
Collapse
Affiliation(s)
- Su-Er W Huskey
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Chun-qi Zhu
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Andreas Fredenhagen
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Jürgen Kühnöl
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Alexandre Luneau
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Zhigang Jian
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Ziping Yang
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Zhuang Miao
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Fan Yang
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Jay P Jain
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Gangadhar Sunkara
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - James B Mangold
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| | - Daniel S Stein
- Drug Metabolism and Pharmacokinetics (S.W.H., C.Z., Z.J., Z.Y., Z.M., J.P.J., G.S., J.B.M) and Translational Medicine (F.Y., D.S.S.), Novartis Institutes for Biomedical Research, East Hanover, New Jersey; and Global Discovery Chemistry, Basel, Switzerland (A.F., J.K., A.L.)
| |
Collapse
|
25
|
Aouri M, Barcelo C, Ternon B, Cavassini M, Anagnostopoulos A, Yerly S, Hugues H, Vernazza P, Günthard HF, Buclin T, Telenti A, Rotger M, Decosterd LA. In Vivo Profiling and Distribution of Known and Novel Phase I and Phase II Metabolites of Efavirenz in Plasma, Urine, and Cerebrospinal Fluid. Drug Metab Dispos 2016; 44:151-61. [PMID: 26553012 DOI: 10.1124/dmd.115.065839] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 11/06/2015] [Indexed: 11/22/2022] Open
Abstract
Efavirenz (EFV) is principally metabolized by CYP2B6 to 8-hydroxy-efavirenz (8OH-EFV) and to a lesser extent by CYP2A6 to 7-hydroxy-efavirenz (7OH-EFV). So far, most metabolite profile analyses have been restricted to 8OH-EFV, 7OH-EFV, and EFV-N-glucuronide, even though these metabolites represent a minor percentage of EFV metabolites present in vivo. We have performed a quantitative phase I and II metabolite profile analysis by tandem mass spectrometry of plasma, cerebrospinal fluid (CSF), and urine samples in 71 human immunodeficiency virus patients taking efavirenz, prior to and after enzymatic (glucuronidase and sulfatase) hydrolysis. We have shown that phase II metabolites constitute the major part of the known circulating efavirenz species in humans. The 8OH-EFV-glucuronide (gln) and 8OH-EFV-sulfate (identified for the first time) in humans were found to be 64- and 7-fold higher than the parent 8OH-EFV, respectively. In individuals (n = 67) genotyped for CYP2B6, 2A6, and CYP3A metabolic pathways, 8OH-EFV/EFV ratios in plasma were an index of CYP2B6 phenotypic activity (P < 0.0001), which was also reflected by phase II metabolites 8OH-EFV-glucuronide/EFV and 8OH-EFV-sulfate/EFV ratios. Neither EFV nor 8OH-EFV, nor any other considered metabolites in plasma were associated with an increased risk of central nervous system (CNS) toxicity. In CSF, 8OH-EFV levels were not influenced by CYP2B6 genotypes and did not predict CNS toxicity. The phase II metabolites 8OH-EFV-gln, 8OH-EFV-sulfate, and 7OH-EFV-gln were present in CSF at 2- to 9-fold higher concentrations than 8OH-EFV. The potential contribution of known and previously unreported EFV metabolites in CSF to the neuropsychological effects of efavirenz needs to be further examined in larger cohort studies.
Collapse
Affiliation(s)
- Manel Aouri
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Catalina Barcelo
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Béatrice Ternon
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Matthias Cavassini
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Alexia Anagnostopoulos
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Sabine Yerly
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Henry Hugues
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Pietro Vernazza
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Huldrych F Günthard
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Thierry Buclin
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Amalio Telenti
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Margalida Rotger
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| | - Laurent A Decosterd
- Laboratory of Clinical Pharmacology, Service of Biomedicine (M.A., B.T., M.R., L.A.D.), Division of Clinical Pharmacology, Service of Biomedicine (M.A., C.B., T.B.), Service of Infectious Diseases (B.T., M.C.), Institute of Microbiology (A.T., M.R.), and Innovation and Development, Service of Biomedicine (H.H.), University Hospital and University of Lausanne, Lausanne, Switzerland; Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland (A.A., H.F.G.); Institute of Medical Virology, University of Zurich, Zurich, Switzerland (H.F.G.); Division of Infectious Diseases, Cantonal Hospital, St. Gallen, Switzerland (P.V.); Laboratory of Virology, University Hospital of Geneva, Geneva, Switzerland (S.Y.); and The J. Craig Venter Institute, La Jolla, California (A.T.)
| |
Collapse
|
26
|
Inhibition of Efavirenz Metabolism by Sertraline and Nortriptyline and Their Effect on Efavirenz Plasma Concentrations. Antimicrob Agents Chemother 2015; 60:1022-8. [PMID: 26643342 DOI: 10.1128/aac.02129-15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/22/2015] [Indexed: 11/20/2022] Open
Abstract
Between 22 and 45% of HIV-positive subjects are likely to report symptoms of depression. Considering this background, a potential pharmacokinetic interaction between the nonnucleoside reverse transcriptase inhibitor efavirenz (EFV) and two antidepressants, sertraline (SRT) and nortriptyline (NT), was studied. Rats were administered EFV alone or together with the antidepressants, and changes in the plasma levels and pharmacokinetic parameters of EFV were analyzed. Additional in vitro experiments with rat and human hepatic microsomes were carried out to evaluate the inhibitory effect of SRT and NT on EFV metabolism by determining the formation rate of the major EFV metabolite (8-OH-EFV). In vivo studies showed similar increases in the plasma levels of EFV when it was coadministered with SRT or NT. However, the studies using rat hepatic microsomes showed a more potent inhibitory effect of NT than of SRT on the metabolism of EFV, with values for the 50% inhibition constant (IC50) and inhibitory constant (Ki) for NT about 9-fold lower than those for SRT. An equation was deduced that explains the similar in vivo effects of SRT and NT in spite of the different in vitro performance data. Using human hepatic microsomes, the strongest inhibitory effect was observed with SRT. In summary, pharmacokinetic interactions between EFV, SRT, and NT, associated with the inhibition of hepatic metabolism of EFV, have been detected in rats. Both antidepressants also inhibit EFV metabolism in human hepatic microsomes, but additional in vivo studies in humans are required to evaluate the clinical implication of this interaction.
Collapse
|
27
|
Apostolova N, Funes HA, Blas-Garcia A, Galindo MJ, Alvarez A, Esplugues JV. Efavirenz and the CNS: what we already know and questions that need to be answered. J Antimicrob Chemother 2015. [PMID: 26203180 DOI: 10.1093/jac/dkv183] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The NNRTI efavirenz has long been one of the most frequently employed antiretroviral drugs in the multidrug regimens used to treat HIV infection, in accordance with its well-demonstrated antiretroviral efficacy and favourable pharmacokinetics. However, growing concern about its adverse effects has sometimes led to efavirenz being replaced by other drugs in the initial treatment selection or to switching of therapy to efavirenz-free regimens in experienced patients. Neurological and neuropsychiatric reactions are the manifestations most frequently experienced by efavirenz-treated patients and range from transitory effects, such as nightmares, dizziness, insomnia, nervousness and lack of concentration, to more severe symptoms including depression, suicidal ideation or even psychosis. In addition, efavirenz has recently been associated with mild/moderate neurocognitive impairment, which is of specific relevance given that half of the patients receiving ART eventually suffer some form of HIV-associated neurocognitive disorder. The mechanisms responsible for efavirenz-induced neurotoxicity are unclear, although growing evidence points to disturbances in brain mitochondrial function and bioenergetics. This review offers a comprehensive overview of the current evidence on the interaction that efavirenz displays with the CNS, including the penetration and concentration of the drug in the brain. We discuss the prevalence, types and specificities of its side effects and recently uncovered cellular mechanisms that may be involved in their development.
Collapse
Affiliation(s)
- Nadezda Apostolova
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain Facultad de Ciencias de la Salud, Universitat Jaume I, Castellón de la Plana, Spain CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Valencia, Spain
| | - Haryes A Funes
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Ana Blas-Garcia
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Valencia, Spain FISABIO-Hospital Universitario Dr Peset, Valencia, Spain
| | - Maria J Galindo
- Unidad de Enfermedades Infecciosas-Medicina Interna, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Angeles Alvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Valencia, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Valencia, Spain FISABIO-Hospital Universitario Dr Peset, Valencia, Spain
| |
Collapse
|
28
|
Frapsauce C, Grabar S, Leruez-ville M, Launay O, Sogni P, Gayet V, Viard J, De Almeida M, Jouannet P, Dulioust E. Impaired sperm motility in HIV-infected men: an unexpected adverse effect of efavirenz? Hum Reprod 2015; 30:1797-806. [DOI: 10.1093/humrep/dev141] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 05/26/2015] [Indexed: 12/25/2022] Open
|
29
|
Central Nervous System Penetration of Antiretroviral Drugs: Pharmacokinetic, Pharmacodynamic and Pharmacogenomic Considerations. Clin Pharmacokinet 2015; 54:581-98. [DOI: 10.1007/s40262-015-0257-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Winston A, Amin J, Clarke A, Else L, Amara A, Owen A, Barber T, Jessen H, Avihingsanon A, Avinghsanon A, Chetchotisakd P, Khoo S, Cooper DA, Emery S, Puls R. Cerebrospinal fluid exposure of efavirenz and its major metabolites when dosed at 400 mg and 600 mg once daily: a randomized controlled trial. Clin Infect Dis 2014; 60:1026-32. [PMID: 25501988 DOI: 10.1093/cid/ciu976] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The optimal penetration of antiretroviral agents into the central nervous system may be a balance between providing adequate drug exposure to inhibit human immunodeficiency virus (HIV) replication while avoiding concentrations associated with neuronal toxicities. METHODS Cerebrospinal fluid (CSF) exposure of efavirenz and the metabolites 7-hydroxy (7OH) and 8-hydroxy (8OH) efavirenz were assessed after at least 12 weeks of therapy in HIV-infected subjects randomized to commence antiretroviral regimens containing efavirenz at either 400 mg or 600 mg once daily. RESULTS Of 28 subjects (14 and 14 on efavirenz 400 mg and 600 mg, respectively), CSF HIV RNA was undetectable in all. Geometric mean CSF efavirenz, 7OH-, and 8OH-efavirenz concentrations (with 90% confidence intervals [CIs]) for the 400-mg and 600-mg dosing groups were 16.5 (13-21) and 19.5 (15-25) ng/mL; 0.6 (.4-.9) and 0.6 (.4-1) ng/mL; and 5.1 (4.0-6.4) and 3.1 (2.1-4.4) ng/mL, respectively. Efavirenz concentration in CSF was >0.51 ng/mL (proposed CSF 50% maximal inhibitory concentration for wild-type virus) in all subjects, and 8OH-efavirenz concentration in CSF was >3.3 ng/mL (a proposed toxicity threshold) in 11 of 14 and 7 of 14 subjects randomized to the 400 mg and 600 mg doses of efavirenz, respectively. Whereas CSF efavirenz concentration was significantly associated with plasma concentration (P < .001) and cytochrome P450 2B6 genotype (CSF efavirenz GG to GT/TT geometric mean ratio, 0.56 [90% CI, .42-.74]), CSF 8OH-efavirenz concentration was not (P = .242 for association and CSF 8OH-efavirenz GG to GT/TT geometric mean ratio, 1.52 [90% CI, .97-2.36]). CONCLUSIONS With both doses of efavirenz studied, CSF concentrations were considered adequate to inhibit HIV replication, although concentrations of 8OH-efavirenz were greater than those reportedly associated with neuronal toxicity. CSF exposure of 8OH-efavirenz was not dependent on plasma exposure and, as we postulate, may be subject to saturable pharmacokinetic effects. CLINICAL TRIALS REGISTRATION NCT01011413.
Collapse
Affiliation(s)
- Alan Winston
- Section of Infectious Diseases, Imperial College London, United Kingdom
| | - Janaki Amin
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Amanda Clarke
- Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Laura Else
- Department of Pharmacology, University of Liverpool
| | - Alieu Amara
- Department of Pharmacology, University of Liverpool
| | - Andrew Owen
- Department of Pharmacology, University of Liverpool
| | - Tristan Barber
- Chelsea and Westminster NHS Foundation Trust, London, United Kingdom
| | | | | | | | | | - Saye Khoo
- Department of Pharmacology, University of Liverpool
| | - David A Cooper
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Sean Emery
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Rebekah Puls
- Kirby Institute, University of New South Wales, Sydney, Australia
| | | | | |
Collapse
|
31
|
Hersman EM, Bumpus NN. A targeted proteomics approach for profiling murine cytochrome P450 expression. J Pharmacol Exp Ther 2014; 349:221-8. [PMID: 24594750 PMCID: PMC3989799 DOI: 10.1124/jpet.113.212456] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/03/2014] [Indexed: 12/23/2022] Open
Abstract
The cytochrome P450 (P450) superfamily of enzymes plays a prominent role in drug metabolism. Although mice are a widely used preclinical model in pharmacology, the expression of murine P450 enzymes at the protein level has yet to be fully defined. Twenty-seven proteins belonging to P450 subfamilies 1A, 2A, 2B, 2C, 2D, 2E, 2F, 2J, 2U, 3A, 4A, 4B, 4F, and 4V were readily detectable in Balb/c mouse tissue using a global mass spectrometry-based proteomics approach. Subsequently, a targeted mass spectrometry-based assay was developed to simultaneously quantify these enzymes in ranges of femtomoles of P450 per microgram of total protein concentration range. This screen was applied to mouse liver microsomes and tissue lysates of kidney, lung, intestine, heart, and brain isolated from mixed-sex fetuses; male and female mice that were 3-4 weeks, 9-10 weeks, and 8-10 months of age; and pregnant mice. CYP1A2 was consistently more abundant in male mouse liver microsomes compared with age-matched females. Hepatic expression of CYP2B9 was more abundant in 3- to 4-week-old male and female mice than in mice of other ages; in addition, CYP2B9 was the only enzyme that was detectable at higher levels in pregnant mouse liver microsomes compared with age-matched females. Interestingly, sexually dimorphic expression of CYP2B9, 2D26, 2E1, and 4B1 was observed in kidney only. The targeted proteomics assay described here can be broadly used as a tool for investigating the expression patterns of P450 enzymes in mice.
Collapse
Affiliation(s)
- Elisabeth M Hersman
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
32
|
Brandmann M, Nehls U, Dringen R. 8-Hydroxy-efavirenz, the primary metabolite of the antiretroviral drug Efavirenz, stimulates the glycolytic flux in cultured rat astrocytes. Neurochem Res 2013; 38:2524-34. [PMID: 24091996 DOI: 10.1007/s11064-013-1165-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 09/25/2013] [Accepted: 09/26/2013] [Indexed: 12/23/2022]
Abstract
In active antiretroviral therapy antiretroviral drugs are employed for the restoration of a functional immune system in patients suffering from the acquired immunodeficiency syndrome. However, potential adverse effects of such compounds to brain cells are discussed in connection with the development of neurocognitive impairments in patients. To investigate potential effects of antiretroviral drugs on cell viability and the glycolytic flux of brain cells, astrocyte-rich primary cultures were exposed to various antiretroviral compounds, including the non-nucleoside reverse transcriptase inhibitor efavirenz. In a concentration of 10 μM, neither efavirenz nor any of the other investigated antiretroviral compounds acutely compromised the cell viability nor altered glucose consumption or lactate production. In contrast, the primary metabolite of efavirenz, 8-hydroxy-efavirenz, stimulated the glycolytic flux in viable astrocytes in a time- and concentration-dependent manner with half-maximal and maximal effects at concentrations of 5 and 10 μM, respectively. The stimulation of glycolytic flux by 8-hydroxy-efavirenz was not additive to that obtained for astrocytes that were treated with the respiratory chain inhibitor rotenone and was abolished by removal of extracellular 8-hydroxy-efavirenz. In a concentration of 10 μM, 8-hydroxy-efavirenz and efavirenz did not affect mitochondrial respiration, while both compounds lowered in a concentration of 60 μM significantly the oxygen consumption by mitochondria that had been isolated form cultured astrocytes, suggesting that the stimulation of glycolytic flux by 8-hydroxy-efavrienz is not caused by direct inhibition of respiration. The observed alteration of astrocytic glucose metabolism by 8-hydroxy-efavirenz could contribute to the adverse neurological side effects reported for patients that are chronically treated with efavirenz-containing medications.
Collapse
Affiliation(s)
- Maria Brandmann
- Centre for Biomolecular Interactions Bremen, University of Bremen, PO. Box 330440, 28334, Bremen, Germany
| | | | | |
Collapse
|