251
|
Natural polymorphisms of human immunodeficiency virus type 1 integrase and inherent susceptibilities to a panel of integrase inhibitors. Antimicrob Agents Chemother 2009; 53:4275-82. [PMID: 19651917 DOI: 10.1128/aac.00397-09] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We evaluated the human immunodeficiency virus type 1 (HIV-1) integrase coding region of the pol gene for the presence of natural polymorphisms in patients during early infection (AHI) and with triple-class drug-resistant HIV-1 (MDR). We analyzed selected recombinant viruses containing patient-derived HIV-1 integrase for susceptibility to a panel of strand transfer integrase inhibitors (InSTI). A pretreatment sequence analysis of the integrase coding region was performed for 112 patients identified during acute or early infection and 15 patients with triple-class resistance. A phenotypic analysis was done on 10 recombinant viruses derived from nine patients against a panel of six diverse InSTI. Few of the polymorphisms associated with in vitro InSTI resistance were identified in the samples from newly infected individuals or those patients with MDR HIV-1. We identified polymorphisms V72I, L74I, T97A, V151I, M154I/L, E157Q, V165I, V201I, I203M, T206S, and S230N. V72I was the most common, seen in 63 (56.3%) of the AHI samples. E157Q was the only naturally occurring mutation thought to contribute to resistance to elvitegravir, raltegravir, and L-870,810. None of the patient-derived viruses demonstrated any significant decrease in susceptibility to the drugs tested. In summary, the integrase coding region contains as much natural variation as that seen in protease, but mutations associated with high-level resistance to existing InSTI are rarely, if ever, present in integrase naïve patients, especially those being used clinically. Most of the highly prevalent polymorphisms have little effect on InSTI susceptibility in the absence of specific primary mutations. Baseline testing for integrase susceptibility in InSTI-naïve patients is not currently warranted.
Collapse
|
252
|
The consequences of HIV infection and antiretroviral therapy use for cardiovascular disease risk: shifting paradigms. Curr Opin HIV AIDS 2009; 4:176-82. [PMID: 19532047 DOI: 10.1097/coh.0b013e328329c62f] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To explore the mechanisms by which HIV infection and antiretroviral therapy (ART) may increase risk for atherosclerotic cardiovascular disease (CVD), with attention to the implications of earlier initiation of ART (i.e. at higher CD4 cell counts than currently recommended by guidelines). RECENT FINDINGS Compared with the general population, HIV-infected patients who receive ART have a greater burden of subclinical and clinical atherosclerotic disease. Findings from a recent international treatment interruption trial (SMART) have redirected attention from ART-related drug toxicity toward a better appreciation for the consequences of untreated HIV infection, which may increase CVD risk through inflammation, upregulation of thrombotic pathways, and ultimately early vascular damage and dysfunction. In addition, CVD risk may increase with some ART, and this risk may be class-specific and/or drug-specific. SUMMARY Compared with untreated HIV, ART may increase or decrease risk of CVD. Reliable data on the relative risk do not exist. A randomized trial of early ART will provide the best data for assessment of the net risks and benefits of ART use on CVD.
Collapse
|
253
|
Abstract
More than 25 years after the licensure of aciclovir and then penciclovir, followed by their respective prodrugs valaciclovir and famciclovir, cases of clinically relevant resistance to these drugs in immunocompetent individuals remain very rare. The aim of this review is to focus on the mechanism of action of these anti HSV drugs and then briefly compare this favourable outcome with that of CMV, HIV, HBV and influenza. A central theme is that resistance is an epiphenomenon of failure to suppress virus replication, so that improved potency and selectivity should be prioritised when developing new drugs rather than activity against resistant strains per se.
Collapse
Affiliation(s)
- Paul D Griffiths
- Centre for Virology, UCL Medical School, Rowland Hill Street, London NW3 2PF, United Kingdom.
| |
Collapse
|
254
|
Gatell JM. [Raltegravir: the first HIV integrase inhibitor. Introduction]. Enferm Infecc Microbiol Clin 2009; 26 Suppl 12:1-2. [PMID: 19572418 DOI: 10.1016/s0213-005x(08)76565-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- José M Gatell
- Servicio de Infecciones, Hospital Clinic, IDIBAPS, Universidad de Barcelona, Barcelona, España.
| |
Collapse
|
255
|
Gatell JM. [Efficacy of raltegravir: from healthy volunteers to phase III trials]. Enferm Infecc Microbiol Clin 2009; 26 Suppl 12:29-33. [PMID: 19572423 DOI: 10.1016/s0213-005x(08)76570-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Raltegravir is the first in a new class of antiretroviral treatments called integrase inhibitors, which work by preventing HIV from inserting its genetic material into the DNA of the human chromosome. Phase I-III studies have shown this drug to have potent antiretroviral action, which is more rapid than that of protease inhibitors. The dose selected in efficacy studies is 400 mg every 12 h. However, due to the favorable pharmacokinetic profile of raltegravir, the possibility of administration of 600 to 800 mg once daily is under study. Given that this drug is not metabolized by the cytochrome P450 system, the potential for pharmacological interactions is low. Moreover, because humans lack a cellular homologue for HIV integrase, raltegravir has a low potential for toxicity. Raltegravir has an intermediate genetic barrier and consequently there may be cross-resistance across the integrase inhibitor class. For all these reasons, raltegravir is an attractive option in treatment-naive and pretreated patients and in those receiving simplification regimens.
Collapse
Affiliation(s)
- José M Gatell
- Servicio de Infecciones, Hospital Clinic, IDIBAPS, Universidad de Barcelona, Barcelona, España.
| |
Collapse
|
256
|
Van Baelen K, Rondelez E, Van Eygen V, Ariën K, Clynhens M, Van den Zegel P, Winters B, Stuyver LJ. A combined genotypic and phenotypic human immunodeficiency virus type 1 recombinant virus assay for the reverse transcriptase and integrase genes. J Virol Methods 2009; 161:231-9. [PMID: 19559730 DOI: 10.1016/j.jviromet.2009.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 06/09/2009] [Accepted: 06/16/2009] [Indexed: 10/20/2022]
Abstract
With the approval of the first HIV-1 integrase inhibitor raltegravir and a second one in phase III clinical development (elvitegravir), genotypic and phenotypic resistance assays are required to guide antiretroviral therapy and to investigate treatment failure. In this study, a genotypic and phenotypic recombinant virus assay was validated for determining resistance against integrase inhibitors. The assays are based on the amplification of a region encompassing not only HIV-1 integrase, but also reverse transcriptase and RNAseH. The overall amplification success was 85% (433/513) and increased to 93% (120/129) for samples with a viral load above 3 log(10) copies/ml. Both B and non-B HIV-1 subtypes could be genotyped successfully (93%; 52/56 and 100%; 49/49, respectively) and reproducibly. The phenotypic assay showed a high success rate (96.5%; 139/144) for subtype B (100%; 19/19) and non-B subtypes (92%; 45/49), and was found to be accurate and reproducible as assessed using well-characterized integrase mutants. Using both assays, baseline resistance to raltegravir and elvitegravir in subtype B and non-B HIV-1 strains selected at random was not observed, although integrase polymorphisms were present at varying prevalence. Biological cutoff values were found to be 2.1 and 2.0 for raltegravir and elvitegravir, respectively. In summary, a genotypic and phenotypic integrase resistance assay was validated successfully for accuracy, reproducibility, analytical and clinical sensitivity, and dynamic range.
Collapse
Affiliation(s)
- Kurt Van Baelen
- Virco BVBA, Generaal De Wittelaan L11 B3, Mechelen, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
257
|
Garvey L, Winston A. Rilpivirine: a novel non-nucleoside reverse transcriptase inhibitor. Expert Opin Investig Drugs 2009; 18:1035-41. [DOI: 10.1517/13543780903055056] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Lucy Garvey
- Clinical Research Fellow, Imperial College London, Division of Medicine, Ground Floor, Clinical Trials, Winston Churchill Wing, London W2 1PG, London, UK ;
- St. Mary's Hospital, Imperial College Healthcare NHS Trust, Department of HIV Medicine, London W2 1NY, UK
| | - Alan Winston
- Clinical Research Fellow, Imperial College London, Division of Medicine, Ground Floor, Clinical Trials, Winston Churchill Wing, London W2 1PG, London, UK ;
- St. Mary's Hospital, Imperial College Healthcare NHS Trust, Department of HIV Medicine, London W2 1NY, UK
| |
Collapse
|
258
|
Rong L, Perelson AS. Modeling HIV persistence, the latent reservoir, and viral blips. J Theor Biol 2009; 260:308-31. [PMID: 19539630 DOI: 10.1016/j.jtbi.2009.06.011] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 04/20/2009] [Accepted: 06/08/2009] [Indexed: 02/02/2023]
Abstract
HIV-1 eradication from infected individuals has not been achieved with the prolonged use of highly active antiretroviral therapy (HAART). The cellular reservoir for HIV-1 in resting memory CD4(+) T cells remains a major obstacle to viral elimination. The reservoir does not decay significantly over long periods of time but is able to release replication-competent HIV-1 upon cell activation. Residual ongoing viral replication may likely occur in many patients because low levels of virus can be detected in plasma by sensitive assays and transient episodes of viremia, or HIV-1 blips, are often observed in patients even with successful viral suppression for many years. Here we review our current knowledge of the factors contributing to viral persistence, the latent reservoir, and blips, and mathematical models developed to explore them and their relationships. We show how mathematical modeling has helped improve our understanding of HIV-1 dynamics in patients on HAART and of the quantitative events underlying HIV-1 latency, reservoir stability, low-level viremic persistence, and emergence of intermittent viral blips. We also discuss treatment implications related to these studies.
Collapse
Affiliation(s)
- Libin Rong
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | | |
Collapse
|
259
|
Suppression of human immunodeficiency virus type 1 (HIV-1) viremia with reverse transcriptase and integrase inhibitors, CD4+ T-cell recovery, and viral rebound upon interruption of therapy in a new model for HIV treatment in the humanized Rag2-/-{gamma}c-/- mouse. J Virol 2009; 83:8254-8. [PMID: 19494021 DOI: 10.1128/jvi.00580-09] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A small animal model that reproduces human immunodeficiency virus type 1 (HIV-1) pathogenesis may allow modeling of new therapeutic strategies in ways not approachable in mononuclear cell culture. We find that, as in humans, combination antiretroviral therapy (ART) in humanized (hu-) Rag2(-/-)gamma(c)(-/-) mice allows suppression of viremia below the limits of detection and recovery of CD4(+) cells, while interruption of ART results in viral rebound and renewed loss of CD4(+) T cells. Failure of ART in infected mice is associated with the appearance of drug resistance mutations. The hu-Rag2(-/-)gamma(c)(-/-) mouse may therefore facilitate testing of novel approaches to HIV replication and persistence.
Collapse
|
260
|
Wanisch K, Yáñez-Muñoz RJ. Integration-deficient lentiviral vectors: a slow coming of age. Mol Ther 2009; 17:1316-32. [PMID: 19491821 DOI: 10.1038/mt.2009.122] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lentiviral vectors are very efficient at transducing dividing and quiescent cells, which makes them highly useful tools for genetic analysis and gene therapy. Traditionally this efficiency was considered dependent on provirus integration in the host cell genome; however, recent results have challenged this view. So called integration-deficient lentiviral vectors (IDLVs) can be produced through the use of integrase mutations that specifically prevent proviral integration, resulting in the generation of increased levels of circular vector episomes in transduced cells. These lentiviral episomes lack replication signals and are gradually lost by dilution in dividing cells, but are stable in quiescent cells. Compared to integrating lentivectors, IDLVs have a greatly reduced risk of causing insertional mutagenesis and a lower risk of generating replication-competent recombinants (RCRs). IDLVs can mediate transient gene expression in proliferating cells, stable expression in nondividing cells in vitro and in vivo, specific immune responses, RNA interference, homologous recombination (gene repair, knock-in, and knock-out), site-specific recombination, and transposition. IDLVs can be converted into replicating episomes, suggesting that if a clinically applicable system can be developed they would also become highly appropriate for stable transduction of proliferating tissues in therapeutic applications.
Collapse
Affiliation(s)
- Klaus Wanisch
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, UK
| | | |
Collapse
|
261
|
Antiretroviral medication adherence and the development of class-specific antiretroviral resistance. AIDS 2009; 23:1035-46. [PMID: 19381075 DOI: 10.1097/qad.0b013e32832ba8ec] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To assess the association between antiretroviral adherence and the development of class-specific antiretroviral medication resistance. DESIGN AND METHODS Literature and conference abstract review of studies assessing the association between adherence to antiretroviral therapy and the development of antiretroviral medication resistance. RESULTS Factors that determine class-specific adherence-resistance relationships include antiretroviral regimen potency, viral fitness or, more specifically, the interplay between the fold-change in resistance and fold-change in fitness caused by drug resistance mutations, and the genetic barrier to antiretroviral resistance. During multidrug therapy, differential drug exposure increases the likelihood of developing resistance. In addition, antiretroviral medications with higher potency and higher genetic barriers to resistance decrease the incidence of resistance for companion antiretroviral medications at all adherence levels. CONCLUSION Knowledge of class-specific adherence-resistance relationships may help clinicians and patients tailor therapy to match individual patterns of adherence in order to minimize the development of resistance at failure. In addition, this information may guide the selection of optimal drug combinations and regimen sequences to improve the durability of antiretroviral therapy.
Collapse
|
262
|
Affiliation(s)
- Jamie D Croxtall
- Wolters Kluwer Health/Adis, 41 Centorian Drive, Mairangi Bay, North Shore 0754, Auckland, New Zealand.
| | | |
Collapse
|
263
|
Chowers MY, Gottesman BS, Leibovici L, Pielmeier U, Andreassen S, Paul M. Reporting of adverse events in randomized controlled trials of highly active antiretroviral therapy: systematic review. J Antimicrob Chemother 2009; 64:239-50. [DOI: 10.1093/jac/dkp191] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
264
|
Pharmacovirological impact of an integrase inhibitor on human immunodeficiency virus type 1 cDNA species in vivo. J Virol 2009; 83:7706-17. [PMID: 19458008 DOI: 10.1128/jvi.00683-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Clinical trials of the first approved integrase inhibitor (INI), raltegravir, have demonstrated a drop in the human immunodeficiency virus type 1 (HIV-1) RNA loads of infected patients that was unexpectedly more rapid than that with a potent reverse transcriptase inhibitor, and apparently dose independent. These clinical outcomes are not understood. In tissue culture, although their inhibition of integration is well documented, the effects of INIs on levels of unintegrated HIV-1 cDNAs have been variable. Furthermore, there has been no report to date on an INI's effect on these episomal species in vivo. Here, we show that prophylactic treatment of transgenic rats with the strand transfer INI GSK501015 reduced levels of viral integrants in the spleen by up to 99.7%. Episomal two-long-terminal-repeat (LTR) circles accumulated up to sevenfold in this secondary lymphoid organ, and this inversely correlated with the impact on the proviral burden. Contrasting raltegravir's dose-ranging study with HIV patients, titration of GSK501015 in HIV-infected animals demonstrated dependence of the INI's antiviral effect on its serum concentration. Furthermore, the in vivo 50% effective concentration calculated from these data best matched GSK501015's in vitro potency when serum protein binding was accounted for. Collectively, this study demonstrates a titratable, antipodal impact of an INI on integrated and episomal HIV-1 cDNAs in vivo. Based on these findings and known biological characteristics of viral episomes, we discuss how integrase inhibition may result in additional indirect antiviral effects that contribute to more rapid HIV-1 decay in HIV/AIDS patients.
Collapse
|
265
|
Abstract
The integrase inhibitor (INI) raltegravir has shown promising results in clinical trials to date, reducing second phase HIV RNA levels by 70% in comparison with standard regimens. These trial results have been limited by the 50 copies/ml detection limit of the HIV RNA assay and have not investigated the effect of an INI regimen on levels of latently infected cells. Mathematical models that duplicated previous raltegravir results were extended to estimate effects of an INI regimen on HIV RNA beyond second phase and on HIV DNA levels. Depending on assumptions underlying later phase HIV RNA generation and its interaction with latently infected cells, HIV RNA in later phases can be lower or show no difference with an INI, and similarly for HIV DNA. If latent infection is maintained by differentiation of stem cells with integrated HIV DNA, then an INI regimen will eventually have no added benefit. Other hypotheses that allow ongoing replication predict continually lower HIV RNA levels with an INI regimen, but this differential effect need not translate to a reduction in latent infection. Investigation of HIV RNA and HIV DNA levels with an INI will provide better understanding of how these components are generated and maintained under antiretroviral therapy.
Collapse
Affiliation(s)
- John M Murray
- School of Mathematics and Statistics, University of New South Wales, Sydney, Australia.
| |
Collapse
|
266
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss recent pharmacological, virological, and clinical data that concern enfuvirtide usage in different antiretroviral combinations. RECENT FINDINGS Randomized, recent trials in multidrug-experienced patients suggest that antiretroviral combinations with enfuvirtide have excellent virological responses with new antiretroviral compounds, including darunavir, etravirine, raltegravir, vicriviroc, and maraviroc. Trials confirm long-term safety, in spite of moderate injection-site reactions or pain, and lack of significant interactions. Preliminary data suggest that switching from enfuvirtide to raltegravir is effective and using enfuvirtide in prophylaxis of mother-to-child transmission is well tolerated. To administer enfuvirtide in an intensification strategy in antiretroviral-naïve or experienced populations may accelerate virological decline. SUMMARY Dosage adaptations to renal insufficiency are not necessary with enfuvirtide. Spinal fluid concentrations and ombilic cord passage are negligible. Durability of virological responses with enfuvirtide in combinations has been confirmed, in spite of injection-site reactions and twice daily subcutaneous administration. Enfuvirtide should be used with at least one other fully active drug in optimized background therapy in multidrug-experienced populations, a possible exception being with entry inhibitors, which may further benefit from the addition of a third active drug. Data concerning enfuvirtide in antiretroviral combinations show accelerated viral load decline, and the possibility of switching from enfuvirtide to raltegravir without modification of optimized background therapy.
Collapse
|
267
|
Effect of rifampin, a potent inducer of drug-metabolizing enzymes, on the pharmacokinetics of raltegravir. Antimicrob Agents Chemother 2009; 53:2852-6. [PMID: 19433563 DOI: 10.1128/aac.01468-08] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Raltegravir is a human immunodeficiency virus type 1 integrase strand transfer inhibitor that is metabolized by glucuronidation via UGT1A1 and may be affected by inducers of UGT1A1, such as rifampin (rifampicin). Two pharmacokinetic studies were performed in healthy subjects: study 1 examined the effect of administration of 600-mg rifampin once daily on the pharmacokinetics of a single dose of 400-mg raltegravir, and study 2 examined the effect of 600-mg rifampin once daily on the pharmacokinetics of 800-mg raltegravir twice daily compared to 400-mg raltegravir twice daily without rifampin. Raltegravir coadministered with rifampin resulted in lower plasma raltegravir concentrations: in study 1, the geometric mean ratios (GMRs) and 90% confidence intervals (90% CIs) for the plasma raltegravir concentration determined 12 h postdose (C(12)), area under the concentration-time curve from 0 h to infinity (AUC(0-infinity)), and maximum concentration of drug in plasma (C(max)) (400-mg raltegravir plus rifampin/400-mg raltegravir) were 0.39 (0.30, 0.51), 0.60 (0.39, 0.91), and 0.62 (0.37, 1.04), respectively. In study 2, the GMRs and 90% CIs for raltegravir C(12), AUC(0-12), and C(max) (800-mg raltegravir plus rifampin/400-mg raltegravir) were 0.47 (0.36, 0.61), 1.27 (0.94, 1.71), and 1.62 (1.12, 2.33), respectively. Doubling the raltegravir dose to 800 mg when coadministered with rifampin therefore compensates for the effect of rifampin on raltegravir exposure (AUC(0-12)) but does not overcome the effect of rifampin on raltegravir trough concentrations (C(12)). Coadministration of rifampin and raltegravir is not contraindicated; however, caution should be used, since raltegravir trough concentrations in the presence of rifampin are likely to be at the lower limit of clinical experience.
Collapse
|
268
|
Katlama C, Murphy R. Emerging role of integrase inhibitors in the management of treatment-experienced patients with HIV infection. Ther Clin Risk Manag 2009; 5:331-40. [PMID: 19536321 PMCID: PMC2697544 DOI: 10.2147/tcrm.s3137] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Integrase is an essential HIV-1-specific enzyme that is an active target for antiretroviral drug development. Recently, a new class of drugs that specifically inhibits strand transfer, one of the three steps of HIV integration into the host DNA, has been developed. Two drugs in this class have reached late stages of development for use in HIV-1 infected individuals: raltegravir, which has just been approved for use in treatment-experienced patients, and elvitegravir, currently in phase III trials. Both are potent with an IC50 in the 30 nM range and active in vitro against wild type as well as in strains highly resistant to all other existing classes of drugs. Clinical trials in both treatment-naïve and -experienced patients have demonstrated raltegravir to be highly effective with an excellent tolerability profile and no specific clinical or metabolic side effects. Longer follow up is necessary to ensure this early safety profile is sustained. The rapid rate of viral decay observed with raltegravir challenges the current understanding of HIV-1 turnover and may open new strategies for long term treatment and management of infected patients.
Collapse
Affiliation(s)
- Christine Katlama
- Pierre et Marie Curie Université - Paris 6, Service de Maladies Infectieuses et Tropicales, Paris, France
| | | |
Collapse
|
269
|
Loizidou EZ, Zeinalipour-Yazdi CD, Christofides T, Kostrikis LG. Analysis of binding parameters of HIV-1 integrase inhibitors: correlates of drug inhibition and resistance. Bioorg Med Chem 2009; 17:4806-18. [PMID: 19450984 DOI: 10.1016/j.bmc.2009.04.058] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 04/07/2009] [Accepted: 04/09/2009] [Indexed: 12/19/2022]
Abstract
This study undertook an exploratory data analysis of the binding parameters of HIV-1 integrase inhibitors. The study group involved inhibitors in preclinical development from the diketo acid, pyrroloquinoline and naphthyridine carboxamide families and the most advanced inhibitors Raltegravir and Elvitegravir. Distinct differences were observed in the energetics of binding between the studied classes of inhibitors that also correlated with drug resistant patterns. Quantitative-property-activity-relationships correlated experimental IC(50) values to the binding energy and the logarithm of the partition coefficient between n-octanol and water (clogP). The approach followed here serves as an improved basis for the development of 'second generation' integrase inhibitors.
Collapse
Affiliation(s)
- Eriketi Z Loizidou
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus.
| | | | | | | |
Collapse
|
270
|
Hazuda D, Iwamoto M, Wenning L. Emerging pharmacology: inhibitors of human immunodeficiency virus integration. Annu Rev Pharmacol Toxicol 2009; 49:377-94. [PMID: 18928385 DOI: 10.1146/annurev.pharmtox.011008.145553] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The first integrase inhibitor licensed to treat HIV-1 infection was approved in late 2007, more than a decade after the introduction of the first inhibitors of the HIV-1 reverse transcriptase and protease. The unique biochemical and molecular mechanism of action of this novel class of antiretroviral drugs is the fundamental basis for their activity in treating multidrug-resistant HIV-1 infection and is important for understanding both the cellular and in vivo pharmacology and metabolism of these agents. In addition, available pharmacokinetic and drug interaction data for raltegravir and elvitegravir, the two integrase inhibitors that are the most advanced in clinical development to date, are reviewed.
Collapse
Affiliation(s)
- Daria Hazuda
- Merck Research Labs, West Point, Pennsylvania 19486, USA.
| | | | | |
Collapse
|
271
|
Chirch LM, Morrison S, Steigbigel RT. Treatment of HIV infection with raltegravir. Expert Opin Pharmacother 2009; 10:1203-11. [DOI: 10.1517/14656560902911488] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
272
|
Abstract
PURPOSE OF REVIEW This article reviews recent studies in the field of clinical pharmacology of antiretroviral drugs and highlights the relevance of the findings to clinical practice in developing countries. RECENT FINDINGS Differences in antiretroviral pharmacokinetics are associated with polymorphisms of genes encoding drug metabolizing enzymes. Inadequate concentrations of antiretrovirals in children are common. A study in African children found subtherapeutic concentrations in 40% of patients receiving efavirenz at recommended doses. SUMMARY Recent findings on the pharmacokinetics of antiretroviral agents relevant to clinical practice in developing countries are reviewed. Widespread poverty impacts negatively on HIV/AIDS treatment and prevention efforts. Improved access to treatment, social and economic support and pharmacology research in target populations are needed.
Collapse
|
273
|
Wittkop L, Breilh D, Da Silva D, Duffau P, Mercié P, Raymond I, Anies G, Fleury H, Saux MC, Dabis F, Fagard C, Thiébaut R, Masquelier B, Pellegrin I. Virological and immunological response in HIV-1-infected patients with multiple treatment failures receiving raltegravir and optimized background therapy, ANRS CO3 Aquitaine Cohort. J Antimicrob Chemother 2009; 63:1251-5. [PMID: 19336453 DOI: 10.1093/jac/dkp114] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The efficacy of raltegravir plus optimized background therapy (OBT) has been demonstrated for antiretroviral (ARV)-experienced HIV-1-infected patients in randomized clinical trials. We studied viro-immunological response, pharmacokinetic parameters and genotypic test results in an observational cohort of multiple ARV class-experienced patients starting a raltegravir-based regimen. METHODS Already enrolled ANRS CO3 Aquitaine Cohort patients with virological failure were included in this study after starting a raltegravir-based regimen (400 mg twice a day, week 0). Virological success was defined by the plasma HIV-1 RNA level [viral load (VL)] <2.7 log(10) copies/mL at week 12 and <1.7 log(10) copies/mL at week 24. One patient was excluded from further analysis (no follow-up after week 4). RESULTS Fifty-one patients [male/female = 43/8, median age = 48 (interquartile range = 43, 55) years] were included. At week 0, median CD4 count was 244 (110; 310)/mm(3) and median VL was 4.2 (3.6, 4.7) log(10) copies/mL. At week 24, 39 (78%) patients experienced virological success: 4 (44%), 14 (82%) and 21 (87%) patients with a genotypic sensitivity score <1, > or =1 and <2 and > or =2 (P = 0.02), respectively. Raltegravir-related mutations emerged in 9 of 11 failing patients (82%): Q148H/R (n = 5), N155S/H (n = 3) and S230N (n = 1). Median CD4 increases from week 0 to week 4 and week 24 were 28 (-4, 85) and 57 (0, 156) cells/mm(3), respectively. A poor immune response was independently associated with a lower VL decline (week 0 to week 12) [odds ratio (OR): 3.5, 95% confidence interval (CI): 1.4, 8.4, for 1 log(10) less] and CD4+% at baseline (OR: 2.6, 95% CI: 0.97, 8.3, for 10% lower). CONCLUSIONS Raltegravir plus OBT provided a good virological success rate in highly pre-treated patients under clinical routine conditions.
Collapse
Affiliation(s)
- Linda Wittkop
- Inserm, Unit Epidemiology and Biostatistics, Bordeaux, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
274
|
Preoperative use of raltegravir-containing regimen as induction therapy: very rapid decline of HIV-1 viral load. AIDS 2009; 23:739. [PMID: 19279447 DOI: 10.1097/qad.0b013e328325a4bd] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
275
|
Wenning LA, Petry AS, Kost JT, Jin B, Breidinger SA, DeLepeleire I, Carlini EJ, Young S, Rushmore T, Wagner F, Lunde NM, Bieberdorf F, Greenberg H, Stone JA, Wagner JA, Iwamoto M. Pharmacokinetics of raltegravir in individuals with UGT1A1 polymorphisms. Clin Pharmacol Ther 2009; 85:623-7. [PMID: 19279563 DOI: 10.1038/clpt.2009.12] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Raltegravir is a human immunodeficiency virus-1 (HIV-1) integrase strand transfer inhibitor metabolized by glucuronidation via UDP-glucuronosyltransferase 1A1 (UGT1A1). In this study, 30 subjects with a UGT1A1*28/*28 genotype (associated with decreased activity of UGT1A1) and 27 UGT1A1*1/*1 control subjects (matched by race, age, gender, and body mass index) received a single 400-mg dose of raltegravir after fasting. No serious adverse experiences were reported, and there were no discontinuations due to adverse experiences. The geometric mean ratio (GMR) (UGT1A1*28/*28 to UGT1A1*1/*1) and 90% confidence interval (CI) were 1.41 (0.96, 2.09) for raltegravir area under the concentration-time curve (AUC(0-infinity)), 1.40 (0.86, 2.28) for maximum plasma concentration (C(max)), and 1.91 (1.43, 2.55) for concentration at the 12-h time point (C(12 h)). No clinically important differences in time to maximum concentration (T(max)) or half-life were observed. Plasma concentrations of raltegravir are modestly higher in individuals with the UGT1A1*28/*28 genotype than in those with the UGT1A1*1/*1 genotype. This increase is not clinically significant, and therefore no dose adjustment of raltegravir is required for individuals with the UGT1A1*28/*28 genotype.
Collapse
Affiliation(s)
- L A Wenning
- Department of Drug Metabolism, Merck Research Laboratories, a division of Merck & Co., Inc., Whitehouse Station, New Jersey, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
276
|
Gupta S, Lataillade M, Farber S, Kozal MJ. Raltegravir with unboosted atazanavir 300 mg twice daily in antiretroviral treatment-experienced participants. ACTA ACUST UNITED AC 2009; 8:87-92. [PMID: 19270153 DOI: 10.1177/1545109709332471] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Raltegravir (RAL) is an HIV integrase inhibitor characterized by potent antiretroviral activity, few adverse effects, and lack of cross-resistance to other antiretroviral (ARV) agents. RAL is emerging as a component of effective alternative ARV therapy for those who experience therapeutic failure or intolerance to reverse transcriptase inhibitors (NRTI and NNRTI) and ritonavir (RTV)-boosted protease inhibitor (PI) containing regimens. The combination of RAL with atazanavir (ATV) without a concomitant NRTI-based backbone or the inclusion of RTV may provide an alternative strategy for those unable to tolerate these latter ARV agents. In this report the authors present a case series of treatment-experienced patients managed with RAL + ATV given without a boosting dose of RTV. All patients tolerated this regimen over a course of 25 to 82 weeks, and had good virologic and immunologic outcome with a decrease in HIV RNA levels to <50 copies/mL and a mean CD4 count increase of 234 cells/mm(3).
Collapse
Affiliation(s)
- Shaili Gupta
- Division of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | |
Collapse
|
277
|
Abstract
Highly active antiretroviral therapy (HAART) has markedly decreased morbidity and mortality in human immunodeficiency virus type 1 (HIV-1)-infected individuals in the developed world. Successful therapy often results in stable plasma levels of HIV-1 RNA below the limits of detection of commercial assays. Nonetheless, HIV-1 has not been cured by HAART. The causes of persistence of HIV infection in the face of current therapy appear to be multifactorial: latent but replication-competent provirus in resting CD4+ T cells, cryptic viral expression below the limits of detection of clinical assays, and viral sanctuary sites might all contribute to persistence. Clearance of HIV infection will almost certainly require a multimodality approach that includes potent suppression of HIV replication, therapies that reach all compartments of residual HIV replication and depletion of any reservoirs of persistent, quiescent proviral infection. This review highlights the basic mechanisms for the establishment and maintenance of viral reservoirs and pharmaceutical approaches towards their elimination.
Collapse
|
278
|
Abstract
BACKGROUND The availability of new classes of antiretroviral drugs has made it possible for HIV-infected individuals who are highly treatment experienced to achieve the goals of immunologic recovery and virologic suppression. Raltegravir is the first integrase inhibitor to be approved by the US Food and Drug Administration for use in antiretroviral treatment- experienced adult patients with viral resistance. OBJECTIVE This article reviews the pharmacology, pharmacokinetics, pharmacodynamics, efficacy, tolerability, resistance profile, drug interactions, and dosing and administration of raltegravir. METHODS Searches of MEDLINE and International Pharmaceutical Abstracts from 1964 to July 2008 were conducted using the terms integrase, raltegravir, and MK-0518. Relevant information was extracted from the identified clinical trials and review articles. Abstracts from the Conference on Retroviruses and Opportunistic Infections (1998-2008); Interscience Conference on Antimicrobial Agents and Chemotherapy (1999-2007); International AIDS Society Conference on HIV Pathogenesis, Treatment and Prevention (2001-2007); and European AIDS Conference (2001-2007) were also searched. RESULTS Raltegravir blocks HIV replication by inhibiting essential strand-transfer activities of integrase. Raltegravir is rapidly absorbed, with a median T(max) of approximately 4 hours in the fasting state. No dose adjustment is recommended in patients with moderate renal or hepatic insufficiency, and raltegravir may be taken without regard to meals. In Phase II studies in treatment-naive patients, raltegravir had efficacy similar to that of standard initial therapies. In 2 interrelated Phase III clinical studies in treatment-experienced patients with drug-resistant disease, the addition of raltegravir to an optimized background regimen significantly lowered HIV RNA compared with optimized background treatment alone (62.1% vs 32.9%, respectively; P < 0.001). Raltegravir was generally well tolerated. The most common adverse effects reported in Phase II/III trials in treatment-experienced patients were diarrhea (16.6%), nausea (9.9%), and headache (9.7%). Cytochrome P450-related drug interactions are not expected, as raltegravir is not a CYP substrate, inducer, or inhibitor. However, to prevent failure of raltegravir, the drug should not be coadministered with rifampin. CONCLUSION Raltegravir is a potent and generally well tolerated antiretroviral agent that may play an important role in the treatment of patients harboring resistance to other antiretrovirals.
Collapse
Affiliation(s)
- Jennifer Cocohoba
- Department of Clinical Pharmacy, School of Pharmacy, University of California San Francisco, San Francisco, California 94143, USA.
| | | |
Collapse
|
279
|
Stellbrink HJ. Raltegravir in the management of HIV-infected patients. DRUG DESIGN DEVELOPMENT AND THERAPY 2009; 2:281-8. [PMID: 19920914 PMCID: PMC2761196 DOI: 10.2147/dddt.s3337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Raltegravir has recently been licensed for the treatment of HIV-1 infection. Currently its use is limited to treatment-experienced patients and subjects with resistant virus. In addition to its activity in the setting of resistance and treatment failure, it appears to have great potential for first-line therapy and as a switch option for subjects with intolerance to other agents, as well. Overall tolerability in clinical trials was excellent, and the toxicity profile is non-overlapping with other agents, with no clear neuropsychiatric, gastrointestinal, or metabolic toxicity. Its metabolization occurs mainly via UGT1A1 rather than by the CYP450 system, resulting in a relatively unproblematic drug interaction profile. The independence of the compound from “boosting” of drug levels with ritonavir is an attractive feature for many patients suffering from ritonavir-associated side effects. However, it has to be dosed twice daily. The unique effect of raltegravir on the establishment of viral latency makes it a logical component of treatment attempts aiming at reducing and controlling this viral sanctuary. This review summarizes the clinical view on the role of this novel compound in HIV therapy.
Collapse
Affiliation(s)
- Hans-Jürgen Stellbrink
- ICH and IPM Study Center Hamburg, Grindelallee 35, Hamburg, Germany. stellbrink@ ich-hamburg.de
| |
Collapse
|
280
|
Sedaghat AR, Siliciano RF, Wilke CO. Constraints on the dominant mechanism for HIV viral dynamics in patients on raltegravir. Antivir Ther 2009. [DOI: 10.1177/135965350901400212] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Raltegravir is the first publicly released HIV integrase inhibitor. In clinical trials, patients on a raltegravir-based highly active antiretroviral therapy (HAART) regimen were observed to have 70% less viraemia in the second-phase decay of viraemia than patients on an efavirenz- based HAART regimen. Because of this accelerated decay of viraemia, raltegravir has been speculated to have greater antiretroviral activity than efavirenz. Alternative explanations for this phenomenon are also possible. For example, the stage in the viral life cycle at which raltegravir acts might explain the distinct viral dynamics produced by this drug. Methods In this report, we use a mathematical model of HIV viral dynamics to explore several hypotheses for why raltegravir causes different viral dynamics than efavirenz. Using the experimentally observed viral dynamics of raltegravir, we calculated constraints on the mechanisms possibly responsible for the unique viral dynamics produced by raltegravir. Results We predicted that the dominant mechanism for the 70% reduction in the second-phase viraemia is not antiviral efficacy but the stage of the HIV viral life cycle at which raltegravir acts. Furthermore, we found that the kinetic constraints placed on the identity of the virus-producing cells of the second phase were most consistent with monocytes/macrophages. Conclusions Our model predictions have important implications for the motivation behind the use of raltegravir and our understanding of the virus-producing cells of the second-phase viraemia. Our results also highlight that the viral dynamics produced by different antiretroviral drugs should not be directly compared with each other.
Collapse
Affiliation(s)
- Ahmad R Sedaghat
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Baltimore, MD, USA
| | - Claus O Wilke
- Section of Integrative Biology, Center for Computational Biology and Bioinformatics, University of Texas at Austin, Austin, TX, USA
- Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
281
|
Katlama C, Tubiana R, Murphy R. Les nouvelles molécules anti-VIH : la place du raltégravir. ACTA ACUST UNITED AC 2009. [DOI: 10.1016/j.antib.2008.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
282
|
Wilson LE, Gallant JE. HIV/AIDS: the management of treatment-experienced HIV-infected patients: new drugs and drug combinations. Clin Infect Dis 2009; 48:214-21. [PMID: 19072245 DOI: 10.1086/595701] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The recent availability of new antiretroviral agents for the treatment of human immunodeficiency virus (HIV) infection has increased treatment options and has improved the durability, tolerability, and long-term efficacy of antiretroviral therapy, even among patients with extensive treatment experience and high levels of drug resistance. This expansion of therapeutic options has led to a revision of current treatment guidelines, which now state that the goal of antiretroviral therapy in all patients is suppression of the plasma HIV RNA level to <50 copies/mL. Successful management of infection for treatment-experienced patients with the new agents requires an understanding of their pharmacology and resistance patterns and the appropriate use of laboratory testing to optimize regimen selection. This review discusses the use of recently approved antiretroviral agents in the management of HIV infection in treatment-experienced patients.
Collapse
Affiliation(s)
- Lucy E Wilson
- Division of Infectious Diseases, Johns Hopkins School of Medicine, 1830 E. Monument Street, Baltimore, MD 21287, USA.
| | | |
Collapse
|
283
|
Radi M, Maga G, Alongi M, Angeli L, Samuele A, Zanoli S, Bellucci L, Tafi A, Casaluce G, Giorgi G, Armand-Ugon M, Gonzalez E, Esté JA, Baltzinger M, Bec G, Dumas P, Ennifar E, Botta M. Discovery of Chiral Cyclopropyl Dihydro-Alkylthio-Benzyl-Oxopyrimidine (S-DABO) Derivatives as Potent HIV-1 Reverse Transcriptase Inhibitors with High Activity Against Clinically Relevant Mutants. J Med Chem 2009; 52:840-51. [DOI: 10.1021/jm801330n] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Marco Radi
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Giovanni Maga
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Maddalena Alongi
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Lucilla Angeli
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Alberta Samuele
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Samantha Zanoli
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Luca Bellucci
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Andrea Tafi
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Gianni Casaluce
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Gianluca Giorgi
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Mercedes Armand-Ugon
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Emmanuel Gonzalez
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - José A. Esté
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Mireille Baltzinger
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Guillaume Bec
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Philippe Dumas
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Eric Ennifar
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| | - Maurizio Botta
- Dipartimento Farmaco Chimico Tecnologico, University of Siena,Via Alcide de Gasperi 2, I-53100 Siena, Italy, Istituto di Genetica Molecolare, IGM-CNR, Via Abbiategrasso 207, I-27100 Pavia, Italy, Dipartimento di Chimica, University of Siena, Via Alcide de Gasperi 2, I-53100 Siena, Italy, Retrovirology Laboratory irsiCaixa, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, E-08916 Badalona, Spain, Architecture et Réactivité de l’ARN, UPR 9002 CNRS/Université Louis Pasteur, 15
| |
Collapse
|
284
|
Makinson A, Reynes J. Raltegravir: first integrase inhibitor for the treatment of HIV infection. Future Virol 2009. [DOI: 10.2217/17460794.4.1.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Raltegravir is the first of the integrase inhibitors to have been approved for treating HIV-1-infected adult patients who have evidence of viral replication and HIV-resistant strains to multiple antiretroviral agents, in combination with other antiretroviral agents. Raltegravir is an inhibitor of strand transfer of reverse-transcribed viral DNA into the host genome, and is administered orally twice daily without boosting. Clinical and pharmacokinetic studies showed strong virological responses, with as yet unmet antiretroviral potency, as well as excellent metabolic tolerance, and a favorable drug drug interaction profile. Further evaluation of possible indications and strategies are needed in terms of long-term tolerability, resistance, penetration in different anatomical compartments, effect on reservoirs, new antiretroviral combinations, first-line treatment, and switching to and treatment intensifications with raltegravir.
Collapse
Affiliation(s)
- Alain Makinson
- Infectious Diseases Department, Montpellier University Hospital, CHU Montpellier, Montpellier, France.
| | - Jacques Reynes
- Infectious Diseases Department, Montpellier University Hospital, CHU Montpellier, Montpellier, France and, HIV infection Research Unit UMR145, University of Montpellier 1, Montpellier, France.
| |
Collapse
|
285
|
Rachlis A, Angel JB, Harris M, Shafran SD, Therrien R, Tremblay C, Wainberg MA. Optimal use of raltegravir (Isentress(R)) in the treatment of HIV-infected adults - Canadian consensus guidelines. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2009; 20:e67-80. [PMID: 20808458 PMCID: PMC2770304 DOI: 10.1155/2009/940745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND OBJECTIVES A meeting of a Canadian group with significant experience and knowledge in HIV management, consisting of five physicians, a pharmacist and an AIDS researcher, was convened. Their goal was to develop guidance for Canadian HIV-treating physicians on the appropriate use of raltegravir (MK-0518, Isentress(R), Merck Frosst Canada Inc) in HIV-infected adults. METHODS Evidence from the published literature and conference presentations, as well as expert opinions of the group members, was considered and evaluated to develop the recommendations. Feedback on the draft recommendations was obtained from this core group, as well as from five other physicians and scientists across Canada with expertise in HIV treatment and antiretroviral drug resistance, and experience in the use of raltegravir. The final recommendations represent the core group's consensus agreement once all feedback was considered. RESULTS/CONCLUSIONS Recommendations were developed to guide physicians in the optimal use of raltegravir. The issues considered included raltegravir's role in overall treatment strategy, efficacy, durability of effect, rate of viral load reduction, resistance, safety/toxicity, pharmacokinetics and drug interactions.
Collapse
Affiliation(s)
- Anita Rachlis
- Division of Infectious Diseases, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto
| | - Jonathan B Angel
- Division of Infectious Diseases, Department of Medicine, Ottawa Hospital and University of Ottawa, Ottawa, Ontario
| | - Marianne Harris
- AIDS Research Program, St. Paul’s Hospital, Vancouver, British Columbia
| | - Stephen D Shafran
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Alberta
| | | | - Cécile Tremblay
- Department of Microbiology and Immunology, Centre Hospitalier de l’Université de Montréal
| | - Mark A Wainberg
- McGill University AIDS Centre, Lady Davis Institute, Jewish General Hospital, Montréal, Quebec
| |
Collapse
|
286
|
Preclinical evaluation of GS-9160, a novel inhibitor of human immunodeficiency virus type 1 integrase. Antimicrob Agents Chemother 2008; 53:1194-203. [PMID: 19104010 DOI: 10.1128/aac.00984-08] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
GS-9160 is a novel and potent inhibitor of human immunodeficiency virus type 1 (HIV-1) integrase (IN) that specifically targets the process of strand transfer. It is an authentic inhibitor of HIV-1 integration, since treatment of infected cells results in an elevation of two-long terminal repeat circles and a decrease of integration junctions. GS-9160 has potent and selective antiviral activity in primary human T lymphocytes producing a 50% effective concentration (EC(50)) of approximately 2 nM, with a selectivity index (50% cytotoxic concentration/EC(50)) of approximately 2,000. The antiviral potency of GS-9160 decreased by 6- to 10-fold in the presence of human serum. The antiviral activity of GS-9160 is synergistic in combination with representatives from three different classes of antiviral drugs, namely HIV-1 protease inhibitors, nonnucleoside reverse transcriptase inhibitors, and nucleotide reverse transcriptase inhibitors. Viral resistance selections performed with GS-9160 yielded a novel pattern of mutations within the catalytic core domain of IN; E92V emerged initially, followed by L74M. While E92V as a single mutant conferred 12-fold resistance against GS-9160, L74M had no effect as a single mutant. Together, these mutations conferred 67-fold resistance to GS-9160, indicating that L74M may potentiate the resistance caused by E92V. The pharmacokinetic profile of GS-9160 in healthy human volunteers revealed that once-daily dosing was not likely to achieve antiviral efficacy; hence, the clinical development of this compound was discontinued.
Collapse
|
287
|
Abstract
BACKGROUND Efavirenz is a potent, safe and tolerable non-nucleoside reverse transcriptase inhibitor (NNRTI) recommended as initial therapy. Recently, several new antiretroviral drugs, including second generation NNRTIs, protease-inhibitors, an integrase-inhibitor and a CCR5 inhibitor, have become or will be shortly available. OBJECTIVE This article will review relevant efficacy and safety data of efavirenz compared to these novel agents or certain common alternate drugs currently used as initial therapy in treatment-naive patients. METHODS Published articles and conference presentations pertaining to efavirenz and/or the newer antiretroviral agents were evaluated. RESULTS/CONCLUSIONS Efavirenz will continue to be preferred initial therapy for now. If longer-term studies of integrase inhibitors and second-generation NNRTIs confirm initial findings, they will eventually supplant efavirenz as preferred first-line agents.
Collapse
Affiliation(s)
- Brookie M Best
- University of California, Division of Pharmacology and Drug Discovery, San Diego, La Jolla, CA 92093-0719, USA.
| | | |
Collapse
|
288
|
|
289
|
|
290
|
Towards novel S-DABOC inhibitors: Synthesis, biological investigation, and molecular modeling studies. Bioorg Med Chem Lett 2008; 18:5777-80. [DOI: 10.1016/j.bmcl.2008.09.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 09/16/2008] [Accepted: 09/17/2008] [Indexed: 11/24/2022]
|
291
|
Sayana S, Khanlou H. Raltegravir: the first in a new class of integrase inhibitors for the treatment of HIV. Expert Rev Anti Infect Ther 2008; 6:419-26. [PMID: 18662108 DOI: 10.1586/14787210.6.4.419] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Raltegravir (formerly known as MK-0518; Isentress) is the first in a new class of integrase inhibitors approved for the use in treatment-experienced adult patients who have evidence of viral replication and HIV strains resistant to multiple antiretroviral agents. It is dosed twice daily with or without food. Raltegravir is a novel antiretroviral that has been shown to be well tolerated. It has demonstrated potent efficacy in the virologic suppression of HIV-1 RNA levels up to 48 weeks in two controlled studies that were conducted in clinically advanced, three-class antiretroviral, treatment-experienced adults.
Collapse
Affiliation(s)
- Shilpa Sayana
- Department of Medicine, AIDS Healthcare Foundation, Los Angeles, CA, USA
| | | |
Collapse
|
292
|
Rational use of antiretroviral therapy in low-income and middle-income countries: optimizing regimen sequencing and switching. AIDS 2008; 22:2053-67. [PMID: 18753937 DOI: 10.1097/qad.0b013e328309520d] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
293
|
Healy EF, Sanders J, King PJ, Robinson WE. A docking study of L-chicoric acid with HIV-1 integrase. J Mol Graph Model 2008; 27:584-9. [PMID: 19004651 DOI: 10.1016/j.jmgm.2008.09.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 09/18/2008] [Accepted: 09/22/2008] [Indexed: 10/21/2022]
Abstract
Human immunodeficiency virus 1 integrase (HIV-1 IN) is the enzyme responsible for integrating the viral DNA into the host genome, and is essential to the replication of the virus. L-Chicoric acid (L-CA) is a bidentate catechol that has been identified as a potent inhibitor of HIV-1 IN. Using the new Autodock 4.0 free-energy function we have obtained a L-CA binding mode that explains its observed potency and is consistent with available experimental data. Because of the alpha,beta-unsaturated ester functionality of the side arms of L-CA we first performed an extensive conformational analysis of L-CA using semiempirical and ab initio calculations. As a result we have identified two distinct L-CA binding modes, one for the s-cis/s-cis and another for the s-cis/s-trans isomers. The most stable conformer was found to be the structure with the alpha,beta-unsaturated ester in the s-cis conformation for both arms of L-CA. This conformer also gave the top-ranked docking solution. Analysis of the interactions with key IN residues, combined with results using a L-CA tetraacetylated derivative and a Q148A IN mutant, correlate well with the experimental data.
Collapse
Affiliation(s)
- Eamonn F Healy
- Department of Chemistry, St. Edward's University, Austin, TX 78704, USA.
| | | | | | | |
Collapse
|
294
|
Moreno Cuerda V, Rubio García R, Barros Aguado C, Morales Conejo M. Nuevos fármacos antirretrovirales: inhibidores del CCR5 y de la integrasa. Rev Clin Esp 2008; 208:463-6. [DOI: 10.1157/13127608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
295
|
|
296
|
Hirsch MS, Günthard HF, Schapiro JM, Brun-Vézinet F, Clotet B, Hammer SM, Johnson VA, Kuritzkes DR, Mellors JW, Pillay D, Yeni PG, Jacobsen DM, Richman DD. Antiretroviral drug resistance testing in adult HIV-1 infection: 2008 recommendations of an International AIDS Society-USA panel. Clin Infect Dis 2008; 47:266-85. [PMID: 18549313 DOI: 10.1086/589297] [Citation(s) in RCA: 350] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Resistance to antiretroviral drugs remains an important limitation to successful human immunodeficiency virus type 1 (HIV-1) therapy. Resistance testing can improve treatment outcomes for infected individuals. The availability of new drugs from various classes, standardization of resistance assays, and the development of viral tropism tests necessitate new guidelines for resistance testing. The International AIDS Society-USA convened a panel of physicians and scientists with expertise in drug-resistant HIV-1, drug management, and patient care to review recently published data and presentations at scientific conferences and to provide updated recommendations. Whenever possible, resistance testing is recommended at the time of HIV infection diagnosis as part of the initial comprehensive patient assessment, as well as in all cases of virologic failure. Tropism testing is recommended whenever the use of chemokine receptor 5 antagonists is contemplated. As the roll out of antiretroviral therapy continues in developing countries, drug resistance monitoring for both subtype B and non-subtype B strains of HIV will become increasingly important.
Collapse
|
297
|
Wenning LA, Friedman EJ, Kost JT, Breidinger SA, Stek JE, Lasseter KC, Gottesdiener KM, Chen J, Teppler H, Wagner JA, Stone JA, Iwamoto M. Lack of a significant drug interaction between raltegravir and tenofovir. Antimicrob Agents Chemother 2008; 52:3253-8. [PMID: 18625763 PMCID: PMC2533504 DOI: 10.1128/aac.00005-08] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 05/09/2008] [Accepted: 07/07/2008] [Indexed: 11/20/2022] Open
Abstract
Raltegravir is a novel human immunodeficiency virus type 1 (HIV-1) integrase inhibitor with potent in vitro activity (95% inhibitory concentration of 31 nM in 50% human serum). This article reports the results of an open-label, sequential, three-period study of healthy subjects. Period 1 involved raltegravir at 400 mg twice daily for 4 days, period 2 involved tenofovir disoproxil fumarate (TDF) at 300 mg once daily for 7 days, and period 3 involved raltegravir at 400 mg twice daily plus TDF at 300 mg once daily for 4 days. Pharmacokinetic profiles were also determined in HIV-1-infected patients dosed with raltegravir monotherapy versus raltegravir in combination with TDF and lamivudine. There was no clinically significant effect of TDF on raltegravir. The raltegravir area under the concentration time curve from 0 to 12 h (AUC(0-12)) and peak plasma drug concentration (C(max)) were modestly increased in healthy subjects (geometric mean ratios [GMRs], 1.49 and 1.64, respectively). There was no substantial effect of TDF on raltegravir concentration at 12 h postdose (C(12)) in healthy subjects (GMR [TDF plus raltegravir-raltegravir alone], 1.03; 90% confidence interval [CI], 0.73 to 1.45), while a modest increase (GMR, 1.42; 90% CI, 0.89 to 2.28) was seen in HIV-1-infected patients. Raltegravir had no substantial effect on tenofovir pharmacokinetics: C(24), AUC, and C(max) GMRs were 0.87, 0.90, and 0.77, respectively. Coadministration of raltegravir and TDF does not change the pharmacokinetics of either drug to a clinically meaningful degree. Raltegravir and TDF may be coadministered without dose adjustments.
Collapse
|
298
|
Affiliation(s)
- Carlo Torti
- Institute of Infectious and Tropical Diseases, University of Brescia, I-25123 Brescia, Italy
| | | |
Collapse
|
299
|
Rhee SY, Liu TF, Kiuchi M, Zioni R, Gifford RJ, Holmes SP, Shafer RW. Natural variation of HIV-1 group M integrase: implications for a new class of antiretroviral inhibitors. Retrovirology 2008; 5:74. [PMID: 18687142 PMCID: PMC2546438 DOI: 10.1186/1742-4690-5-74] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2008] [Accepted: 08/07/2008] [Indexed: 11/10/2022] Open
Abstract
HIV-1 integrase is the third enzymatic target of antiretroviral (ARV) therapy. However, few data have been published on the distribution of naturally occurring amino acid variation in this enzyme. We therefore characterized the distribution of integrase variants among more than 1,800 published group M HIV-1 isolates from more than 1,500 integrase inhibitor (INI)-naïve individuals. Polymorphism rates equal or above 0.5% were found for 34% of the central core domain positions, 42% of the C-terminal domain positions, and 50% of the N-terminal domain positions. Among 727 ARV-naïve individuals in whom the complete pol gene was sequenced, integrase displayed significantly decreased inter- and intra-subtype diversity and a lower Shannon's entropy than protease or RT. All primary INI-resistance mutations with the exception of E157Q--which was present in 1.1% of sequences--were nonpolymorphic. Several accessory INI-resistance mutations including L74M, T97A, V151I, G163R, and S230N were also polymorphic with polymorphism rates ranging between 0.5% to 2.0%.
Collapse
Affiliation(s)
- Soo-Yon Rhee
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Tommy F Liu
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mark Kiuchi
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Rafael Zioni
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Robert J Gifford
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Robert W Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
300
|
Brown TT. Approach to the human immunodeficiency virus-infected patient with lipodystrophy. J Clin Endocrinol Metab 2008; 93:2937-45. [PMID: 18685115 PMCID: PMC2515075 DOI: 10.1210/jc.2008-1019] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Accepted: 06/23/2008] [Indexed: 01/16/2023]
Abstract
Subcutaneous atrophy and central fat accumulation are common among HIV-infected patients receiving highly active antiretroviral therapy, and may be accompanied by dyslipidemia and insulin resistance. These fat changes, although commonly referred to together as lipodystrophy, are best considered as separate disorders, with distinct pathogeneses and treatment approaches. These morphological and metabolic abnormalities first appeared after introduction of protease inhibitors more than 10 yr ago, but research has demonstrated that their pathogenesis is multifactorial, with contributions from other antiretroviral medications, patient-related factors, and HIV itself. Switching to a less toxic highly active antiretroviral therapy regimen has shown partial effectiveness for the management of fat atrophy and lipid abnormalities. Lifestyle modification or surgical approaches are the treatment of choice for lipohypertrophy, although novel therapies targeting the GH axis show promise. HIV-related dyslipidemia may be difficult to treat, and can be complicated by drug-drug interactions between some lipid-lowering medications and antiretroviral medications. Treatment of diabetes in HIV-infected patients should generally follow established guidelines, but thiazolidinediones, rather than metformin, may be considered first-line treatment in a patient with lipoatrophy, given their potential to increase sc fat. The contribution of body fat changes and metabolic abnormalities to cardiovascular risk and the changing risk profiles of newer antiretroviral regimens are under intense investigation.
Collapse
Affiliation(s)
- Todd T Brown
- Division of Endocrinology and Metabolism, Johns Hopkins University, 1830 East Monument Street, Suite 333, Baltimore, Maryland 21287, USA.
| |
Collapse
|