1
|
Thu HHK, Schemelev AN, Ostankova YV, Davydenko VS, Reingardt DE, Tran T, Thanh LC, Truong TXL, Totolian AA. Experience in Diagnostic of HIV Drug Resistance in the Mekong Delta Region, Vietnam: A Comparative Analysis Before and After the COVID-19 Pandemic. Diagnostics (Basel) 2025; 15:1279. [PMID: 40428272 PMCID: PMC12109980 DOI: 10.3390/diagnostics15101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Vietnam has made significant strides in reducing the prevalence of HIV infection and achievements in its antiretroviral treatment program. However, the COVID-19 pandemic and financial challenges in the healthcare system have posed significant obstacles to maintaining effective HIV treatment and monitoring, particularly among vulnerable populations. This study aims to evaluate the situation of HIV drug resistance among patients who have experienced treatment failure in the Mekong Delta region and to compare data from 2019 to 2022. Methods: The study material was blood plasma samples from HIV-infected individuals with ART failure: 316 collected in 2019 and 326 collected in 2022. HIV-1 genotyping and mutation detection were performed based on an analysis of the nucleotide sequences of the Pol gene region. A total of 116 HIV-infected individuals with virological failure in 2019 and 2022 were assessed for HIV drug resistance. Results: The study revealed a high proportion of participants with viral loads exceeding 1000 copies/mL, significantly increasing from 12.0% in 2019 to 23.9% in 2022 (OR = 2.3; p = 0.0001). HIV drug resistance mutations were detected in 84.21% of cases in 2019 and 92.59% in 2022. The prevalence of concurrent resistance to NRTIs and NNRTIs was 37.5% and 30.13% in 2019 and 2022, respectively. There was a statistically significant decrease in NNRTI resistance (OR = 0.32, χ2 = 5.43, p < 0.05). In contrast, multi-drug resistance to protease inhibitors rose from 18.52% to 45.21% (φ* = 0.00403, p < 0.05). Triple-class resistance was identified only in 2022 (17.81%). The most common mutations included M184I/V, D67N, K103N, Y181C, and V82A/S/T, with D67N rising significantly from 3.13% to 21.92%. The predominant subtype was CRF01_AE. Conclusion: A high prevalence of viral non-suppression and HIV drug resistance was observed among patients in the Mekong Delta region, particularly after the onset of the COVID-19 pandemic. Our study highlights the ongoing challenges that the HIV/AIDS treatment program in Vietnam must address in the post-pandemic period to sustain its success and achieve the goals of the country's HIV prevention strategies.
Collapse
Affiliation(s)
- Huynh Hoang Khanh Thu
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 70000, Vietnam; (H.H.K.T.); (T.T.); (L.C.T.)
| | - Alexandr N. Schemelev
- Saint Petersburg Pasteur Institute, St. Petersburg 197101, Russia; (Y.V.O.); (V.S.D.); (D.E.R.); (A.A.T.)
| | - Yulia V. Ostankova
- Saint Petersburg Pasteur Institute, St. Petersburg 197101, Russia; (Y.V.O.); (V.S.D.); (D.E.R.); (A.A.T.)
| | - Vladimir S. Davydenko
- Saint Petersburg Pasteur Institute, St. Petersburg 197101, Russia; (Y.V.O.); (V.S.D.); (D.E.R.); (A.A.T.)
| | - Diana E. Reingardt
- Saint Petersburg Pasteur Institute, St. Petersburg 197101, Russia; (Y.V.O.); (V.S.D.); (D.E.R.); (A.A.T.)
| | - Ton Tran
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 70000, Vietnam; (H.H.K.T.); (T.T.); (L.C.T.)
| | - Le Chi Thanh
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 70000, Vietnam; (H.H.K.T.); (T.T.); (L.C.T.)
| | | | - Areg A. Totolian
- Saint Petersburg Pasteur Institute, St. Petersburg 197101, Russia; (Y.V.O.); (V.S.D.); (D.E.R.); (A.A.T.)
| |
Collapse
|
2
|
Garland JM, Mayan H, Kantor R. Treatment of Advanced HIV in the Modern Era. Drugs 2025:10.1007/s40265-025-02181-1. [PMID: 40354016 DOI: 10.1007/s40265-025-02181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 05/14/2025]
Abstract
Antiretroviral therapy has transformed human immunodeficiency virus (HIV) infection from a fatal illness into a manageable chronic condition. However, despite remarkable progress, the HIV epidemic remains a global health challenge, with ambitious targets such as 95-95-95 by 2030 at risk of being unmet. While antiretroviral therapy availability has expanded worldwide, gaps persist, including unawareness of HIV status, inconsistent medication uptake, and limited engagement in care across diverse settings. Advanced HIV represents a particularly challenging yet underexplored aspect of HIV care. Its definition is complex, complicating efforts to address the needs of this vulnerable population. This review characterizes advanced HIV populations, defines them by spectra of immune suppression, antiretroviral therapy exposure, and drug resistance, and explores contemporary approaches to their management, with a particular focus on drug resistance and its clinical implications in modern HIV care. It highlights the unique challenges faced by individuals presenting late to care, those with limited care engagement, and aging populations with long-term exposure to HIV and antiretroviral therapy. By defining these populations, refining our understanding of advanced HIV, and addressing the diverse needs of affected individuals, providers can enhance outcomes and develop strategies to overcome barriers to care. Bridging these critical gaps is essential to advancing global efforts to end the HIV epidemic, both in the USA and worldwide.
Collapse
Affiliation(s)
- Joseph M Garland
- The Miriam Hospital, Providence, RI, USA
- Brown University, Providence, RI, USA
| | - Haim Mayan
- Sheba Medical Center, Tel Aviv, Israel
- Tel Aviv University, Tel Aviv, Israel
| | - Rami Kantor
- The Miriam Hospital, Providence, RI, USA.
- Brown University, Providence, RI, USA.
| |
Collapse
|
3
|
Aung S, Novitsky V, Steingrimsson J, Gillani FS, Howison M, Nagel K, Solomon M, Bertrand T, Bhattarai L, Fulton J, Bandy U, Kantor R. Acquired Human Immunodeficiency Virus Type 1 Drug Resistance in Rhode Island, USA, 2004-2021. J Infect Dis 2024; 230:1422-1433. [PMID: 39041648 PMCID: PMC11646588 DOI: 10.1093/infdis/jiae344] [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: 04/22/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) acquired drug resistance (ADR) compromises antiretroviral therapy (ART). METHODS We aggregated all HIV-1 protease-reverse transcriptase-integrase sequences over 2004-2021 at the largest HIV center in Rhode Island and evaluated ADR extent, trends, and impact using Stanford Database tools. Trends were measured with Mann-Kendall statistic, and multivariable regressions evaluated resistance predictors. RESULTS Sequences were available for 914 ART-experienced persons. Overall ADR to any drug decreased from 77% to 49% (-0.66 Mann-Kendall statistic); nucleoside reverse transcriptase inhibitors 65% to 32%, nonnucleoside reverse transcriptase inhibitors 53% to 43%, and protease inhibitors 28% to 7% (2004-2021), and integrase strand transfer inhibitors 16% to 13% (2017-2021). Multiclass resistance decreased from 44% to 12% (2-class) and 12% to 6% (3-class). In 2021, 94% had at least one 3-drug or 2-drug one-pill-once-daily (OPOD) option. Males and those exposed to more ART regimens were more likely to have ≥2-class resistance, and higher regimen exposure was also associated with fewer OPOD options. CONCLUSIONS Comprehensive analyses within a densely-sampled HIV epidemic over 2004-2021 demonstrated decreasing ADR. Continued ADR monitoring is important to maintain ART success, particularly with rising INSTI use in all lines of therapy and 2-drug and long-acting formulations.
Collapse
Affiliation(s)
- Su Aung
- Department of Medicine, University of California, San Francisco
| | | | | | | | | | | | | | | | - Lila Bhattarai
- Rhode Island Department of Health, Providence, Rhode Island
| | | | - Utpala Bandy
- Rhode Island Department of Health, Providence, Rhode Island
| | | |
Collapse
|
4
|
Nasreddine R, Darcis G, Yombi JC, De Munter P, Florence E, Van Praet J, Demeester R, Allard SD, Schroeder M, Dusabineza AC, Delforge M, De Wit S. A characterization of the HIV population with limited/exhausted treatment options: a multicenter Belgian study. Acta Clin Belg 2024; 79:153-159. [PMID: 38813800 DOI: 10.1080/17843286.2024.2359184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVE Describe the prevalence and characteristics of people living with HIV (PLWH) in Belgium with limited/exhausted treatment options. METHODS A cross-sectional, multicenter study involving adult treatment-experienced individuals with limited/exhausted treatment options defined as having a multi-drug resistant HIV-1 or a history of multiple treatment changes. The primary outcome was to determine the prevalence of these individuals and classify them based on their two most recent consecutive HIV-1 viral loads (VLs): suppressed (2 VLs < 50 copies/mL), intermediate (≥1 VL between 50-200 copies/mL), or unsuppressed (2 VLs > 200 copies/mL). Secondary outcome was to characterize the participants included in this analysis. RESULTS There were 119 individuals included (prevalence of 0.97%; 119 of 12 282 in care). The majority were aged > 50 years (88.2%), women represented 35.3%, and individuals were primarily White (54.7%). Median (IQR) CD4+ T-cell count was 635 (400-875) cells/µL and most (42%) were on a 3-drug ART regimen. Overall, 87.4% were classified as suppressed, 9.2% as intermediate, and 3.4% as unsuppressed. On multivariable analysis, CD4+ T-cell count < 200 cells/µL was associated with being classified as intermediate or unsuppressed (p = 0.004). CONCLUSION In this analysis of PLWH in Belgium, individuals with limited/exhausted treatment options represented a small fraction. Most were on a 3-drug ART regimen, were virologically suppressed, and had a CD4+ T-cell count within normal range. A small proportion were not virologically suppressed while others, despite being suppressed, were on ≥ 4-drug ART regimens. As such, new therapeutic options are needed to achieve and maintain virologic suppression in such individuals while decreasing their pill burden.
Collapse
Affiliation(s)
- Rakan Nasreddine
- Department of Infectious Diseases, Saint-Pierre University Hospital, Brussels, Belgium
| | - Gilles Darcis
- Department of Infectious Diseases, Liège University Hospital, Liège, Belgium
| | - Jean Cyr Yombi
- Department of Internal Medicine and Infectious Diseases, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Paul De Munter
- Department of General Internal Medicine, Leuven University Hospital, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Eric Florence
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Jens Van Praet
- Department of Nephrology and Infectious Diseases, AZ Sint-Jan Brugge-Oostende, Brugge, Belgium
| | - Rémy Demeester
- Department of Internal Medicine and Infectious Diseases, University Hospital of Charleroi, Lodelinsart, Belgium
| | - Sabine D Allard
- Department of Internal Medicine and Infectious Diseases, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | | | - Marc Delforge
- Department of Infectious Diseases, Saint-Pierre University Hospital, Brussels, Belgium
| | - Stéphane De Wit
- Department of Infectious Diseases, Saint-Pierre University Hospital, Brussels, Belgium
| |
Collapse
|
5
|
Carr A, Mackie NE, Paredes R, Ruxrungtham K. HIV drug resistance in the era of contemporary antiretroviral therapy: A clinical perspective. Antivir Ther 2023; 28:13596535231201162. [PMID: 37749751 DOI: 10.1177/13596535231201162] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Contemporary antiretroviral therapy (ART) regimens have high barriers to the development of drug resistance. However, resistance to earlier antiretrovirals and uncommon cases of resistance to contemporary ART illustrate the continued need for good clinical management of HIV drug resistance. Here, we describe HIV drug-resistance mechanisms, the interaction of HIV drug-resistant mutations and the patterns of drug resistance to contemporary ART. We then provide guidance on the management of HIV drug resistance, including how to limit the development of resistance and manage virologic failure that is complicated by resistance. To complement this, links to resources and treatment guidelines are provided that can assist with the interpretation of HIV drug resistance test results and optimal ART selection in the clinic.
Collapse
Affiliation(s)
- Andrew Carr
- HIV and Immunology Unit, St Vincent's Hospital, Sydney, NSW, Australia
- University of New South Wales, Sydney, NSW, Australia
| | | | - Roger Paredes
- Department of Infectious Diseases, Hospital Germans Trias i Pujol, Barcelona, Spain
- IrsiCaixa AIDS Research Institute, Barcelona, Spain
| | - Kiat Ruxrungtham
- Chula Vaccine Research Center (Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- The HIV Netherlands Australia Thailand Research Collaboration (HIV-NAT), Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| |
Collapse
|
6
|
Ostankova YV, Shchemelev AN, Thu HHK, Davydenko VS, Reingardt DE, Serikova EN, Zueva EB, Totolian AA. HIV Drug Resistance Mutations and Subtype Profiles among Pregnant Women of Ho Chi Minh City, South Vietnam. Viruses 2023; 15:2008. [PMID: 37896785 PMCID: PMC10612098 DOI: 10.3390/v15102008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
According to the latest data released by UNAIDS, the global number of people living with HIV (PLHIV) in 2021 was 38.4 million, with 1.5 million new HIV infections. In different countries, a significant proportion of these cases occur in the adult fertile population aged 15-49 years. According to UNAIDS, Vietnam had a national HIV prevalence of 0.3% of the total population at the end of 2019, with approximately 230,000 PLHIV. The most effective way to prevent mother-to-child transmission of HIV is ART to reduce maternal viral load. HIV-infected pregnant women should undergo monthly monitoring, especially before the expected date of delivery. The aim of our work was to analyze subtypic structure and drug-resistant variants of HIV in pregnant women in Ho Chi Minh City. The study material was blood plasma samples from HIV-infected pregnant women: 31 women showed virological failure of ART, and 30 women had not previously received therapy. HIV-1 genotyping and mutation detection were performed based on analysis of the nucleotide sequences of the pol gene region. More than 98% of sequences genotyped as HIV-1 sub-subtype CRF01_AE. When assessing the occurrence of drug resistance mutations, genetic resistance to any drug was detected in 74.41% (95% CI: 62.71-85.54%) of patients. These included resistance mutations to protease inhibitors in 60.66% (95% CI: 47.31-72.93%) of patients, to NRTIs in 8.20% (95% CI: 2.72-18.10%), and to NNRTIs in 44.26% (95% CI: 31.55-57.52%). Mutations associated with NRTI (2) and NNRTI (8) resistance as well as PI mutations (12), including minor ones, were identified. The high prevalence of drug resistance mutations found in this study among pregnant women, both in therapeutically naive individuals and in patients with virological failure of ART, indicates that currently used regimens in Vietnam are insufficient to prevent vertical HIV infection.
Collapse
Affiliation(s)
- Yulia V. Ostankova
- Saint Petersburg Pasteur Institute, 19710 St. Petersburg, Russia; (Y.V.O.)
| | | | | | | | - Diana E. Reingardt
- Saint Petersburg Pasteur Institute, 19710 St. Petersburg, Russia; (Y.V.O.)
| | - Elena N. Serikova
- Saint Petersburg Pasteur Institute, 19710 St. Petersburg, Russia; (Y.V.O.)
| | - Elena B. Zueva
- Saint Petersburg Pasteur Institute, 19710 St. Petersburg, Russia; (Y.V.O.)
| | - Areg A. Totolian
- Saint Petersburg Pasteur Institute, 19710 St. Petersburg, Russia; (Y.V.O.)
| |
Collapse
|
7
|
Jaha B, Schenkel CD, Jörimann L, Huber M, Zaheri M, Neumann K, Leemann C, Calmy A, Cavassini M, Kouyos RD, Günthard HF, Metzner KJ. Prevalence of HIV-1 drug resistance mutations in proviral DNA in the Swiss HIV Cohort Study, a retrospective study from 1995 to 2018. J Antimicrob Chemother 2023; 78:2323-2334. [PMID: 37545164 PMCID: PMC10477134 DOI: 10.1093/jac/dkad240] [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: 04/26/2023] [Accepted: 07/19/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Genotypic resistance testing (GRT) is routinely performed upon diagnosis of HIV-1 infection or during virological failure using plasma viral RNA. An alternative source for GRT could be cellular HIV-1 DNA. OBJECTIVES A substantial number of participants in the Swiss HIV Cohort Study (SHCS) never received GRT. We applied a method that enables access to the near full-length proviral HIV-1 genome without requiring detectable viraemia. METHODS Nine hundred and sixty-two PBMC specimens were received. Our two-step nested PCR protocol was applied to generate two overlapping long-range amplicons of the HIV-1 genome, sequenced by next-generation sequencing (NGS) and analysed by MinVar, a pipeline to detect drug resistance mutations (DRMs). RESULTS Six hundred and eighty-one (70.8%) of the samples were successfully amplified, sequenced and analysed by MinVar. Only partial information of the pol gene was contained in 82/681 (12%), probably due to naturally occurring deletions in the proviral sequence. All common HIV-1 subtypes were successfully sequenced. We detected at least one major DRM at high frequency (≥15%) in 331/599 (55.3%) individuals. Excluding APOBEC-signature (G-to-A mutation) DRMs, 145/599 (24.2%) individuals carried at least one major DRM. RT-inhibitor DRMs were most prevalent. The experienced time on ART was significantly longer in DRM carriers (P = 0.001) independent of inclusion or exclusion of APOBEC-signature DRMs. CONCLUSIONS We successfully applied a reliable and efficient method to analyse near full-length HIV-1 proviral DNA and investigated DRMs in individuals with undetectable or low viraemia. Additionally, our data underscore the need for new computational tools to exclude APOBEC-related hypermutated NGS sequence reads for reporting DRMs.
Collapse
Affiliation(s)
- Bashkim Jaha
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Corinne D Schenkel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Lisa Jörimann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Maryam Zaheri
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Kathrin Neumann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Christine Leemann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | - Matthias Cavassini
- Division of Infectious Diseases, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Roger D Kouyos
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Huldrych F Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Karin J Metzner
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
8
|
Zhukova A, Dunn D, Gascuel O. Modeling Drug Resistance Emergence and Transmission in HIV-1 in the UK. Viruses 2023; 15:1244. [PMID: 37376544 DOI: 10.3390/v15061244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
A deeper understanding of HIV-1 transmission and drug resistance mechanisms can lead to improvements in current treatment policies. However, the rates at which HIV-1 drug resistance mutations (DRMs) are acquired and which transmitted DRMs persist are multi-factorial and vary considerably between different mutations. We develop a method for the estimation of drug resistance acquisition and transmission patterns. The method uses maximum likelihood ancestral character reconstruction informed by treatment roll-out dates and allows for the analysis of very large datasets. We apply our method to transmission trees reconstructed on the data obtained from the UK HIV Drug Resistance Database to make predictions for known DRMs. Our results show important differences between DRMs, in particular between polymorphic and non-polymorphic DRMs and between the B and C subtypes. Our estimates of reversion times, based on a very large number of sequences, are compatible but more accurate than those already available in the literature, with narrower confidence intervals. We consistently find that large resistance clusters are associated with polymorphic DRMs and DRMs with long loss times, which require special surveillance. As in other high-income countries (e.g., Switzerland), the prevalence of sequences with DRMs is decreasing, but among these, the fraction of transmitted resistance is clearly increasing compared to the fraction of acquired resistance mutations. All this indicates that efforts to monitor these mutations and the emergence of resistance clusters in the population must be maintained in the long term.
Collapse
Affiliation(s)
- Anna Zhukova
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - David Dunn
- UK MRC Clinical Trials Unit, University College London, London WC1V 6LJ, UK
| | - Olivier Gascuel
- Institut de Systématique, Evolution, Biodiversité (ISYEB)-URM 7205 CNRS, Muséum National d'Histoire Naturelle, SU, EPHE & UA, 75005 Paris, France
| |
Collapse
|
9
|
Thoueille P, Seybold U, Decosterd LA, Desfontaine V. Development and validation of a liquid chromatography coupled to tandem mass spectrometry method for the monitoring of temsavir plasma concentrations in people living with HIV. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1214:123575. [PMID: 36542988 DOI: 10.1016/j.jchromb.2022.123575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/08/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
A majority of people living with HIV (PLWH) now have access to HIV treatment with high antiviral potency and favorable tolerability profile. However, in some treatment experienced PLWH viral strains resistant to major current classes of antiretrovirals have emerged, usually due to periods with continued virus replication in the presence of failing drug regimens and thus selection pressure. In such context, new treatment options are therefore needed. Fostemsavir (RUKOBIA®) is the prodrug of temsavir, a first-in-class oral attachment inhibitor approved for the treatment of heavily treatment-experienced adults with multidrug-resistant HIV-1 infection. In this case RUKOBIA® is part of a complex regimen of antiretroviral drugs, often in addition to other drugs for chronic co-morbidities (e.g., heart disease, diabetes mellitus, hepatic and renal impairment, etc). In such a multi-drug regimen context, therapeutic drug monitoring (TDM) of temsavir can be necessary to exclude or adjust for relevant drug-drug interactions. A highly selective assay by liquid chromatography method coupled to tandem mass spectrometry (LC-MS/MS) was therefore developed for the quantification of temsavir in human plasma. A convenient sample preparation using protein precipitation with acetonitrile followed by supernatant dilution was carried out. Temsavir and fostemsavir were separated in less than 2 min using a multi-step UPLC gradient, thus ensuring adequate quantification of temsavir. The assay for the quantification of temsavir was extensively validated over the large range of clinically relevant concentrations from 1 to 10,000 ng/mL, in accordance with international bioanalytical method guidelines. The method achieves excellent performance in terms of trueness (99.7 - 105.3%), repeatability and intermediate precision (both from 1.6% to 5.8%). This LC-MS/MS method is now part of the routine analyses of the Laboratory of the Service of Clinical Pharmacology of Lausanne (CHUV), Switzerland, as an integrated part of our general TDM Service for antiretrovirals.
Collapse
Affiliation(s)
- Paul Thoueille
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Ulrich Seybold
- Med. Klinik und Poliklinik IV, LMU Klinikum, Ludwig-Maximilians-University, Munich, Germany
| | - Laurent A Decosterd
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Vincent Desfontaine
- Service and Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Chen J, Chen H, Li J, Luo L, Kang R, Liang S, Zhu Q, Lu H, Zhu J, Shen Z, Feng Y, Liao L, Xing H, Shao Y, Ruan Y, Lan G. Genetic network analysis of human immunodeficiency virus sexual transmission in rural Southwest China after the expansion of antiretroviral therapy: A population-based study. Front Microbiol 2022; 13:962477. [PMID: 36060743 PMCID: PMC9434148 DOI: 10.3389/fmicb.2022.962477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/26/2022] [Indexed: 11/28/2022] Open
Abstract
Background This study is used to analyze the genetic network of HIV sexual transmission in rural areas of Southwest China after expanding antiretroviral therapy (ART) and to investigate the factors associated with HIV sexual transmission through the genetic network. Materials and methods This was a longitudinal genetic network study in Guangxi, China. The baseline survey and follow-up study were conducted among patients with HIV in 2015, and among those newly diagnosed from 2016 to 2018, respectively. A generalized estimating equation model was employed to explore the factors associated with HIV transmission through the genetic linkage between newly diagnosed patients with HIV (2016-2018) and those at baseline (2015-2017), respectively. Results Of 3,259 identified HIV patient sequences, 2,714 patients were at baseline, and 545 were newly diagnosed patients with HIV at follow-up. A total of 8,691 baseline objectives were observed by repeated measurement analysis. The prevention efficacy in HIV transmission for treated HIV patients was 33% [adjusted odds ratio (AOR): 0.67, 95% confidence interval (CI): 0.48-0.93]. Stratified analyses indicated the prevention efficacy in HIV transmission for treated HIV patients with a viral load (VL) of <50 copies/ml and those treated for 4 years with a VL of <50 copies/ml to be 41 [AOR: 0.59, 95% CI: 0.43-0.82] and 65% [AOR: 0.35, 95% CI: 0.24-0.50], respectively. No significant reduction in HIV transmission occurred among treated HIV patients with VL missing or treated HIV patients on dropout. Some factors were associated with HIV transmission, including over 50 years old, men, Zhuang and other nationalities, with less than secondary schooling, working as a farmer, and heterosexual transmission. Conclusion This study reveals the role of ART in reducing HIV transmission, and those older male farmers with less than secondary schooling are at high risk of HIV infection at a population level. Improvements to ART efficacy for patients with HIV and precision intervention on high-risk individuals during the expansion of ART are urgently required.
Collapse
Affiliation(s)
- Jin Chen
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for Acquired Immune Deficiency Syndrome (AIDS)/Sexually Transmitted Disease (STD) Control and Prevention, Chinese Center for Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Huanhuan Chen
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Jianjun Li
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Liuhong Luo
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Ruihua Kang
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for Acquired Immune Deficiency Syndrome (AIDS)/Sexually Transmitted Disease (STD) Control and Prevention, Chinese Center for Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Shujia Liang
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Qiuying Zhu
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Huaxiang Lu
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Jinhui Zhu
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Zhiyong Shen
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| | - Yi Feng
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for Acquired Immune Deficiency Syndrome (AIDS)/Sexually Transmitted Disease (STD) Control and Prevention, Chinese Center for Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Lingjie Liao
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for Acquired Immune Deficiency Syndrome (AIDS)/Sexually Transmitted Disease (STD) Control and Prevention, Chinese Center for Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Hui Xing
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for Acquired Immune Deficiency Syndrome (AIDS)/Sexually Transmitted Disease (STD) Control and Prevention, Chinese Center for Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Yiming Shao
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for Acquired Immune Deficiency Syndrome (AIDS)/Sexually Transmitted Disease (STD) Control and Prevention, Chinese Center for Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Yuhua Ruan
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for Acquired Immune Deficiency Syndrome (AIDS)/Sexually Transmitted Disease (STD) Control and Prevention, Chinese Center for Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Guanghua Lan
- Guangxi Key Laboratory of Major Infectious Disease Prevention Control and Biosafety Emergency Response, Guangxi Center for Disease Control and Prevention, Nanning, China
| |
Collapse
|
11
|
D'Antoni ML, Andreatta K, Acosta R, Martin H, Chang S, Martin R, White KL. Brief Report: Bictegravir/Emtricitabine/Tenofovir Alafenamide Efficacy in Participants With Preexisting Primary Integrase Inhibitor Resistance Through 48 Weeks of Phase 3 Clinical Trials. J Acquir Immune Defic Syndr 2022; 89:433-440. [PMID: 34897227 PMCID: PMC8860220 DOI: 10.1097/qai.0000000000002888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/15/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Preexisting drug resistance limits the utility of HIV antiretroviral therapy. Studies have demonstrated safety and efficacy of bictegravir/emtricitabine/tenofovir alafenamide (B/F/TAF), including in patients with M184V/I substitutions. SETTING We investigated virologic outcomes through 48 weeks of B/F/TAF treatment in individuals with preexisting primary integrase strand transfer inhibitor resistance (INSTI-R). METHODS Preexisting INSTI-R was retrospectively evaluated from 7 B/F/TAF studies. INSTI-R was assessed by historical genotypes and/or baseline RNA or DNA sequencing. Viral loads were measured at all visits. RESULTS Preexisting primary INSTI-R substitutions were detected in 20 of the 1907 participants (1.0%). The 20 participants were predominantly male (75%), were Black (65%), had HIV-1 subtype B (85%), and had baseline median CD4 counts of 594 cells/mm3 and median age of 52 years. Most of the participants (n = 19) were virologically suppressed at baseline and had one primary INSTI-R substitution, E92G, Y143C/H, S147G, Q148H/K/R, N155S, or R263K, +/-secondary substitutions. All suppressed participants maintained virologic suppression throughout 48 weeks without any viral blips. One treatment-naive participant had virus with Q148H+G140S that was fully sensitive to bictegravir but only partially to dolutegravir (phenotype <2.5-fold change and >4-fold change, respectively). With a baseline viral load of 30,000 copies/mL, this participant was virologically suppressed by week 4 and maintained <50 copies/mL through week 48. CONCLUSIONS This small cohort with primary INSTI-R achieved and/or maintained virologic suppression through 48 weeks of B/F/TAF treatment. Consistent with the potent in vitro activity of bictegravir against most INSTI-R patterns, B/F/TAF may be a potential treatment option for patients with select preexisting INSTI-R, if confirmed by further studies.
Collapse
|
12
|
Balaji S, Madhumathi J, Bhargava A, Singh T, Mahajan N, Ambalkar D, Aggarwal S. Patterns of human immunodeficiency virus drug resistance mutations in people living with human immunodeficiency virus in India: A scoping review. Indian J Sex Transm Dis AIDS 2022; 43:13-19. [PMID: 35846552 PMCID: PMC9282713 DOI: 10.4103/ijstd.ijstd_2_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 11/04/2022] Open
Abstract
The human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) epidemic still exists as a major global public health burden, especially in the middle- and low-income countries. Antiretroviral therapy (ART) remains a sole option to reduce the mortality and morbidity associated with this disease as no approved vaccine candidates are available. About 67% of the people living with HIV (PLHIV) have received the ART in 2019 worldwide. As a consequence of increased ART regimes, the prevalence of drug resistance mutations (DRM) also has been escalating and it would become a significant barrier in achieving the United Nations Programme on HIV/AIDS goal of eliminating HIV by 2030. So far, nucleoside reverse transcriptase inhibitors (NRTI), non-nucleoside reverse transcriptase inhibitors (NNRTI), and protease inhibitor-(PI) associated DRM have been reported across the globe with a considerable escalation in the annual prevalence rate of pretreatment NNRTI DRM. Conversely, NRTI-associated DRM is still under 5%, with a few scattered reports of significant increase from few countries such as southern and eastern Africa. Likewise, in India, the propositions of NRTI and NNRTI-associated DRM have increased since the commencement of the nationwide ART program in 2004. In agreement to the global trend, M1841/V, a type of NNRTI, remains as a dominant DRM among PLHIV. In this review, we tried to collate various mechanisms of DRM in PLHIV. In addition, patterns of HIV DRM in India and their future challenges on drug-related mutations have been discussed.
Collapse
Affiliation(s)
- Sivaraman Balaji
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research-Headquarters, New Delhi, India
| | - J. Madhumathi
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research-Headquarters, New Delhi, India
| | - Aradhana Bhargava
- Apex Regional STD Centre, VMMC and Safdarjung Hospital, New Delhi, India
| | - Tanvi Singh
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research-Headquarters, New Delhi, India
| | - Nupur Mahajan
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research-Headquarters, New Delhi, India
| | - Deepti Ambalkar
- Department of Lab Medicine, Max Super Specialty Hospital, New Delhi, India
| | - Sumit Aggarwal
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research-Headquarters, New Delhi, India
| |
Collapse
|
13
|
McComsey GA, Lingohr-Smith M, Rogers R, Lin J, Donga P. Real-World Adherence to Antiretroviral Therapy Among HIV-1 Patients Across the United States. Adv Ther 2021; 38:4961-4974. [PMID: 34390465 PMCID: PMC8363866 DOI: 10.1007/s12325-021-01883-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/03/2021] [Indexed: 01/19/2023]
Abstract
Introduction Recent changes in antiretroviral therapies (ARTs) may have affected medication adherence of people living with human immunodeficiency virus-1 (HIV-1). In this study adherence to ART regimens among patients with HIV-1 (PWH) across the US during a recent time period was examined and study findings were stratified by US region and state. Methods A retrospective observational study using the Symphony Health Solution Integrated Dataverse database was conducted. Patients ≥ 18 years of age who had a diagnosis of HIV-1 (without an HIV-2 diagnosis) and who were treated with ART between July 2017 and September 2018 (first pharmacy record: index date) were selected from the data source. Both patients who had not been previously treated with ART and those who were treatment experienced were included. Patients were required to have ≥ 1 medical/pharmacy record ≥ 12 months after their index date (follow-up period). Patient characteristics were examined during a 12-month pre-index period. During the follow-up, medication adherence, measured as the proportion of days covered (PDC), was examined for all patients and stratified by US region and state. Results Among 206,474 adult PWH treated with ART, mean age was 47.9 years, 73.4% were male, and 30.0% were Caucasian. The most prevalent comorbid conditions were hyperlipidemia (25.1%), depressive disorders (14.8%), and type 2 diabetes (12.1%). During the follow-up period, mean (standard deviation) PDC was 74.1% (25.9%) among PWH across the US [Midwest: 74.4% (25.5%); Northeast: 74.3% (26.1%); South: 73.2% (26.3%); West: 76.4% (24.8%)]. Across all US regions, > 60% of PWH had adherence < 90% and > 40% had adherence < 80%; the West had the highest adherent population. Conclusions Among PWH treated with ART across the US, a majority had suboptimal adherence. Implementation of strategies to improve ART adherence, including clinical consideration of ARTs with high genetic barriers to resistance, is needed in the US. Supplementary Information The online version contains supplementary material available at 10.1007/s12325-021-01883-8.
Collapse
Affiliation(s)
- Grace A. McComsey
- University Hospitals of Cleveland and Case Western Reserve University, Cleveland, OH USA
| | | | | | - Jay Lin
- Novosys Health, Green Brook, NJ USA
| | - Prina Donga
- Janssen Scientific Affairs LLC, Titusville, NJ USA
| |
Collapse
|
14
|
Scherrer AU, Traytel A, Braun DL, Calmy A, Battegay M, Cavassini M, Furrer H, Schmid P, Bernasconi E, Stoeckle M, Kahlert C, Trkola A, Kouyos RD, Tarr P, Marzolini C, Wandeler G, Fellay J, Bucher H, Yerly S, Suter F, Hirsch H, Huber M, Dollenmaier G, Perreau M, Martinetti G, Rauch A, Günthard HF. Cohort Profile Update: The Swiss HIV Cohort Study (SHCS). Int J Epidemiol 2021; 51:33-34j. [PMID: 34363666 DOI: 10.1093/ije/dyab141] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alexandra U Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Anna Traytel
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Dominique L Braun
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | - Manuel Battegay
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Matthias Cavassini
- Division of Infectious Diseases, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Hansjakob Furrer
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Patrick Schmid
- Division of Infectious Diseases, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Enos Bernasconi
- Division of Infectious Diseases, Regional Hospital Lugano, Lugano, Switzerland
| | - Marcel Stoeckle
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Christian Kahlert
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St Gallen, St Gallen, Switzerland.,Division of Infectious Diseases and Hospital Epidemiology, Children's Hospital of Eastern Switzerland, St Gallen, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Roger D Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Philip Tarr
- University Department of Medicine, Kantonsspital Bruderholz, University of Basel, Bruderholz, Switzerland
| | - Catia Marzolini
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Gilles Wandeler
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jacques Fellay
- Precision Medicine Unit, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Heiner Bucher
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sabine Yerly
- Division of Infectious Diseases and Laboratory of Virology, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | - Franziska Suter
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans Hirsch
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | | | - Matthieu Perreau
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Gladys Martinetti
- Department of Microbiology, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Andri Rauch
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
15
|
Armenia D, Di Carlo D, Flandre P, Bouba Y, Borghi V, Forbici F, Bertoli A, Gori C, Fabeni L, Gennari W, Pinnetti C, Mondi A, Cicalini S, Gagliardini R, Vergori A, Bellagamba R, Malagnino V, Montella F, Colafigli M, Latini A, Marocco R, Licthner M, Andreoni M, Mussini C, Ceccherini-Silberstein F, Antinori A, Perno CF, Santoro MM. HIV MDR is still a relevant issue despite its dramatic drop over the years. J Antimicrob Chemother 2021; 75:1301-1310. [PMID: 31976521 DOI: 10.1093/jac/dkz554] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/16/2019] [Accepted: 12/11/2019] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES To evaluate the prevalence and therapeutic relevance of drug resistance among isolates from ART-experienced HIV-1-infected patients over the past two decades in Italy. METHODS Dynamics of resistance to one, two and three or more antiretroviral classes were evaluated from 1999-2018. Virological success (VS) after the latest therapy switch was evaluated according to cumulative class resistance and cumulative genotypic susceptibility score (Stanford HIV_DB algorithm). RESULTS Among 13 663 isolates (from 6739 patients), resistance to at least one drug class decreased sharply from 1999 to 2010 (≤2001, 84.6%; 2010, 43.6%; P < 0.001), then remained relatively constant at ∼40% during 2010-18, with the proportion of resistance to three or more classes also stable (∼5%). After 2008, integrase inhibitor resistance slightly increased from 5.6% to 9.7% in 2018 and contributed to resistance, particularly in isolates with resistance to three or more classes (one class, 8.4%; two classes, 15.3%; three or more classes, 34.7%, P < 0.001). Among 1827 failing patients with an available follow-up, by 1 year after genotype-guided therapy start the probability of VS was 87.6%. Patients with cumulative resistance to three or more classes and receiving a poorly active regimen showed the lowest probability (62.6%) of VS (P < 0.001) compared with all other patients (≥81.8%). By Cox regression analysis, cumulative MDR and receiving poorly active antiretroviral regimens were associated with a lower hazard of VS compared with those without resistance. CONCLUSIONS A dramatic drop of HIV-1 drug resistance at failure has been achieved over the last two decades in Italy; resistance to three or more classes is low but present among currently failing patients. Its management still requires a rational and careful diagnostic and therapeutic approach.
Collapse
Affiliation(s)
- D Armenia
- UniCamillus International University of Health and Medical Sciences, Rome, Italy.,University of Rome 'Tor Vergata', Department of Experimental Medicine, Rome, Italy
| | - D Di Carlo
- University of Milan, Pediatric Clinical Research Center 'Romeo and Enrica Invernizzi', Milan, Italy
| | - P Flandre
- INSERM, Sorbonne Université, Institut Pierre Louis d'Epidémiologie et de Santé Publique (IPLESP) équipe Epidémiologie clinique des maladies virales chroniques, Paris, France
| | - Y Bouba
- University of Rome 'Tor Vergata', Department of Experimental Medicine, Rome, Italy.,Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management (CIRCB), Yaoundé, Cameroon
| | - V Borghi
- Clinic of Infectious Diseases, University Hospital, University of Modena and Reggio Emilia, Modena, Italy
| | - F Forbici
- Laboratory of Virology, National Institute for Infectious Diseases 'Lazzaro Spallanzani', IRCCS, Rome, Italy
| | - A Bertoli
- University of Rome 'Tor Vergata', Department of Experimental Medicine, Rome, Italy
| | - C Gori
- Laboratory of Virology, National Institute for Infectious Diseases 'Lazzaro Spallanzani', IRCCS, Rome, Italy
| | - L Fabeni
- University of Rome 'Tor Vergata', Department of Experimental Medicine, Rome, Italy
| | - W Gennari
- Microbiology Unit, University Hospital of Modena, Modena, Italy
| | - C Pinnetti
- HIV/AIDS Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani' IRCCS, Rome, Italy
| | - A Mondi
- HIV/AIDS Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani' IRCCS, Rome, Italy
| | - S Cicalini
- HIV/AIDS Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani' IRCCS, Rome, Italy
| | - R Gagliardini
- HIV/AIDS Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani' IRCCS, Rome, Italy
| | - A Vergori
- HIV/AIDS Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani' IRCCS, Rome, Italy
| | - R Bellagamba
- HIV/AIDS Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani' IRCCS, Rome, Italy
| | - V Malagnino
- Clinical Infectious Diseases, University Hospital 'Tor Vergata', Rome, Italy
| | - F Montella
- Infectious disease Unit, San Giovanni Addolorata Hospital, Rome, Italy
| | - M Colafigli
- Unit of Dermatology and Sexually Transmitted Diseases, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - A Latini
- Unit of Dermatology and Sexually Transmitted Diseases, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - R Marocco
- Infectious Diseases Unit, 'Sapienza' University, Polo Pontino, Latina, Italy
| | - M Licthner
- Infectious Diseases Unit, 'Sapienza' University, Polo Pontino, Latina, Italy
| | - M Andreoni
- Clinical Infectious Diseases, University Hospital 'Tor Vergata', Rome, Italy
| | - C Mussini
- Clinic of Infectious Diseases, University Hospital, University of Modena and Reggio Emilia, Modena, Italy
| | | | - A Antinori
- HIV/AIDS Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani' IRCCS, Rome, Italy
| | - C F Perno
- Laboratory of Virology, National Institute for Infectious Diseases 'Lazzaro Spallanzani', IRCCS, Rome, Italy
| | - M M Santoro
- University of Rome 'Tor Vergata', Department of Experimental Medicine, Rome, Italy
| |
Collapse
|
16
|
Raffi F, Gaultier A, Pozniak A, Molina JM, Jessen H, Antinori A, Soria A, Cavellec M, Le Thuaut A, Ningre M, de Wit S. Five-year follow-up of patients enrolled in the NEAT 001/ANRS 143 randomized clinical trial: NEAT 001/ANRS 143 LONG TERM study. J Antimicrob Chemother 2021; 75:1618-1622. [PMID: 32211883 DOI: 10.1093/jac/dkaa056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/18/2020] [Accepted: 01/29/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Few long-term data are available in subjects having initiated ART with an NRTI-sparing regimen. OBJECTIVES Outcomes of subjects enrolled in the NEAT 001/ANRS 143 randomized clinical trial (comparing ritonavir-boosted darunavir + raltegravir versus ritonavir-boosted darunavir + tenofovir disoproxil fumarate/emtricitabine) were retrospectively collected, through anonymized electronic case report forms, up to 6 years post-enrolment. METHODS The last NEAT 001 visit (Week 96) was conducted in 745/805 randomized subjects (363/401 ritonavir-boosted darunavir + raltegravir and 382/404 ritonavir-boosted darunavir + tenofovir disoproxil fumarate/emtricitabine). Of these, 430 were enrolled in NEAT 001/ANRS 143 LONG TERM (NLT) study (201 raltegravir, 229 tenofovir disoproxil fumarate/emtricitabine), with a median follow-up of 44.4 months. RESULTS During NLT follow-up, the proportion of AIDS, non-AIDS events, virological rebound and serious adverse events, discontinuation for virological failure and for adverse events did not differ between groups; discontinuations for virological failure since NEAT 001 inclusion were more frequent in subjects with baseline CD4 <200 cells/mm3 (11.9% versus 5.3%; P = 0.077). At last follow-up, a quarter of subjects (22.2% for ritonavir-boosted darunavir + raltegravir and 29.7% for ritonavir-boosted darunavir + tenofovir disoproxil fumarate/emtricitabine) were still receiving their initial regimen. Integrase inhibitor exposure was not associated with weight gain (P = 0.48), while tenofovir disoproxil fumarate exposure was associated with a trend to higher creatinine increase (P = 0.067). CONCLUSIONS After a median of 5.6 years, subjects initiating ritonavir-boosted darunavir + raltegravir or ritonavir-boosted darunavir + tenofovir disoproxil fumarate/emtricitabine experienced few serious clinical adverse events. Most discontinuations were for reasons unrelated to adverse events or virological failure.
Collapse
Affiliation(s)
- François Raffi
- INSERM CIC 1413 Nantes University, and Service des Maladies Infectieuses, Centre hospitalier universitaire de l'Hôtel-Dieu, Nantes, France
| | | | - Anton Pozniak
- Chelsea and Westminster Hospital NHS Foundation Trust and LSHTM, London, UK
| | - Jean-Michel Molina
- Department of Infectious Diseases, Saint-Louis hospital, 1 avenue C. Vellefaux, Paris, and Assistance Publique Hopitaux de Paris and University of Paris Diderot, Sorbonne Paris Cité, France
| | - Heiko Jessen
- Gemeinschaftspraxis Jessen-Stein, Berlin, Germany
| | - Andrea Antinori
- Viral Immunodeficiencies Unit, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Albane Soria
- INSERM CIC 1413 Nantes University, and Service des Maladies Infectieuses, Centre hospitalier universitaire de l'Hôtel-Dieu, Nantes, France
| | - Morane Cavellec
- INSERM CIC 1413 Nantes University, and Service des Maladies Infectieuses, Centre hospitalier universitaire de l'Hôtel-Dieu, Nantes, France
| | | | - Maelle Ningre
- CHU de Nantes, Direction de la Recherche, Nantes, France
| | - Stéphane de Wit
- Division of Infectious Diseases, Saint Pierre University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
17
|
|
18
|
Nguyen H, Thorball CW, Fellay J, Böni J, Yerly S, Perreau M, Hirsch HH, Kusejko K, Thurnheer MC, Battegay M, Cavassini M, Kahlert CR, Bernasconi E, Günthard HF, Kouyos RD, The Swiss HIV Cohort Study. Systematic screening of viral and human genetic variation identifies antiretroviral resistance and immune escape link. eLife 2021; 10:e67388. [PMID: 34061023 PMCID: PMC8169104 DOI: 10.7554/elife.67388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022] Open
Abstract
Background Considering the remaining threat of drug-resistantmutations (DRMs) to antiretroviral treatment (ART) efficacy, we investigated how the selective pressure of human leukocyte antigen (HLA)-restricted cytotoxic T lymphocytes drives certain DRMs' emergence and retention. Methods We systematically screened DRM:HLA class I allele combinations in 3997 ART-naïve Swiss HIV Cohort Study (SHCS) patients. For each pair, a logistic regression model preliminarily tested for an association with the DRM as the outcome. The three HLA:DRM pairs remaining after multiple testing adjustment were analyzed in three ways: cross-sectional logistic regression models to determine any HLA/infection time interaction, survival analyses to examine if HLA type correlated with developing specific DRMs, and via NetMHCpan to find epitope binding evidence of immune escape. Results Only one pair, RT-E138:HLA-B18, exhibited a significant interaction between infection duration and HLA. The survival analyses predicted two pairs with an increased hazard of developing DRMs: RT-E138:HLA-B18 and RT-V179:HLA-B35. RT-E138:HLA-B18 exhibited the greatest significance in both analyses (interaction term odds ratio [OR] 1.169 [95% confidence interval (CI) 1.075-1.273]; p-value<0.001; survival hazard ratio 12.211 [95% CI 3.523-42.318]; p-value<0.001). The same two pairs were also predicted by netMHCpan to have epitopic binding. Conclusions We identified DRM:HLA pairs where HLA presence is associated with the presence or emergence of the DRM, indicating that the selective pressure for these mutations alternates direction depending on the presence of these HLA alleles. Funding Funded by the Swiss National Science Foundation within the framework of the SHCS, and the University of Zurich, University Research Priority Program: Evolution in Action: From Genomes Ecosystems, in Switzerland.
Collapse
Affiliation(s)
- Huyen Nguyen
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of ZurichZurichSwitzerland
- Institute of Medical Virology, Swiss National Center for Retroviruses, University of ZurichZurichSwitzerland
| | - Christian Wandell Thorball
- School of Life Sciences, École PolytechniqueFédérale de LausanneSwitzerland
- Precision Medicine Unit, Lausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Jacques Fellay
- School of Life Sciences, École PolytechniqueFédérale de LausanneSwitzerland
- Precision Medicine Unit, Lausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Jürg Böni
- Institute of Medical Virology, Swiss National Center for Retroviruses, University of ZurichZurichSwitzerland
| | - Sabine Yerly
- Laboratory of Virology, Geneva University Hospital, University of GenevaGenevaSwitzerland
| | - Matthieu Perreau
- Division of Immunology and Allergy, University Hospital Lausanne, University of LausanneLausanneSwitzerland
| | - Hans H Hirsch
- Transplantation & Clinical Virology, Department of Biomedicine, University of BaselBaselSwitzerland
- Infectious Diseases and Hospital Epidemiology, Department of Medicine, University Hospital BaselBaselSwitzerland
- Clinical Virology, Laboratory Medicine, University Hospital BaselBaselSwitzerland
| | - Katharina Kusejko
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of ZurichZurichSwitzerland
- Institute of Medical Virology, Swiss National Center for Retroviruses, University of ZurichZurichSwitzerland
| | - Maria Christine Thurnheer
- University Clinic of Infectious Diseases, University Hospital of Bern, University of BernBernSwitzerland
| | - Manuel Battegay
- Infectious Diseases and Hospital Epidemiology, Department of Medicine, University Hospital BaselBaselSwitzerland
| | - Matthias Cavassini
- Department of Infectious Diseases, Centre Hospitalier Universitaire Vaudois, University of LausanneLausanneSwitzerland
| | - Christian R Kahlert
- Division of Infectious Diseases and Hospital Epidemiology, Kantonsspital St. GallenSt. GallenSwitzerland
| | - Enos Bernasconi
- Division of Infectious Diseases, Regional HospitalLuganoSwitzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of ZurichZurichSwitzerland
- Institute of Medical Virology, Swiss National Center for Retroviruses, University of ZurichZurichSwitzerland
| | - Roger D Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of ZurichZurichSwitzerland
- Institute of Medical Virology, Swiss National Center for Retroviruses, University of ZurichZurichSwitzerland
| | | |
Collapse
|
19
|
Glass TR, Günthard HF, Calmy A, Bernasconi E, Scherrer AU, Battegay M, Steffen A, Böni J, Yerly S, Klimkait T, Cavassini M, Furrer H. The Role of Human Immunodeficiency Virus (HIV) Asymptomatic Status When Starting Antiretroviral Therapy on Adherence and Treatment Outcomes and Implications for Test and Treat: The Swiss HIV Cohort Study. Clin Infect Dis 2021; 72:1413-1421. [PMID: 32157270 DOI: 10.1093/cid/ciaa239] [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: 11/25/2019] [Accepted: 03/09/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Since the advent of universal test-and-treat , more people living with human immunodeficiency virus (PLHIV) initiating antiretroviral therapy (ART) are asymptomatic with a preserved immune system. We explored the impact of asymptomatic status on adherence and clinical outcomes. METHODS PLHIV registered in the Swiss HIV Cohort Study (SHCS) between 2003 and 2018 were included. We defined asymptomatic as Centers for Disease Control and Prevention stage A within 30 days of starting ART, non-adherence as any self-reported missed doses and viral failure as two consecutive viral load>50 copies/mL after >24 weeks on ART. Using logistic regression models, we measured variables associated with asymptomatic status and adherence and Cox proportional hazard models to assess association between symptom status and viral failure. RESULTS Of 7131 PLHIV, 76% started ART when asymptomatic and 1478 (22%) experienced viral failure after a median of 1.9 years (interquartile range, 1.1-4.2). In multivariable models, asymptomatic PLHIV were more likely to be younger, men who have sex with men, better educated, have unprotected sex, have a HIV-positive partner, have a lower viral load, and have started ART more recently. Asymptomatic status was not associated with nonadherence (odds ratio, 1.03 [95% confidence interval {CI}, .93-1.15]). Asymptomatic PLHIV were at a decreased risk of viral failure (adjusted hazard ratio, 0.87 [95% CI, .76-1.00]) and less likely to develop resistance (14% vs 27%, P < .001) than symptomatic PLHIV. CONCLUSIONS Despite concerns regarding lack of readiness, our study found no evidence of adherence issues or worse clinical outcomes in asymptomatic PLHIV starting ART.
Collapse
Affiliation(s)
- Tracy R Glass
- Department of Medicine, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, University Hospital Geneva, Geneva, Switzerland
| | - Enos Bernasconi
- Division of Infectious Diseases, Regional Hospital Lugano, Lugano, Switzerland
| | | | - Manuel Battegay
- University of Basel, Basel, Switzerland.,Division of Infectious Diseases, University Hospital Basel, Basel, Switzerland
| | - Ana Steffen
- Division of Infectious Diseases, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Jürg Böni
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Sabine Yerly
- Laboratory of Virology, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Thomas Klimkait
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Matthias Cavassini
- Division of Infectious Diseases, University Hospital Lausanne, Lausanne, Switzerland
| | - Hansjakob Furrer
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Kusejko K, Salazar-Vizcaya L, Braun DL, Tarr PE, Bernasconi E, Doco-Lecompte T, Cavassini M, Schmid P, Du Pasquier R, Hauser C, Günthard HF, Kouyos RD. Self-reported Neurocognitive Impairment in People Living With Human Immunodeficiency Virus (HIV): Characterizing Clusters of Patients With Similar Changes in Self-reported Neurocognitive Impairment, 2013-2017, in the Swiss HIV Cohort Study. Clin Infect Dis 2021; 71:637-644. [PMID: 31504323 DOI: 10.1093/cid/ciz868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 09/01/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Self-reported neurocognitive impairment (SRNI) in people living with human immunodeficiency virus type 1 (HIV-1) infection is frequent. We use longitudinal information on SRNI in the Swiss HIV Cohort Study (SHCS) to identify and characterize groups of patients with persisting SRNI over time. METHODS We included all SHCS patients who were assessed for SRNI during at least 5 visits spanning at least 2.5 years in 2013-2017. We first compared patients with SRNI to those without SRNI over the whole study period. Second, we used a hierarchical cluster algorithm to identify groups of patients with similar changes of SRNI over time. In both analyses, we studied clinical and demographic factors potentially influencing SRNI. RESULTS In total, 79 683 questionnaires of 11 029 patients contained information about SRNI, and 8545 of 11 029 (77.5%) patients had longitudinal information. The overall percentage of patients with SRNI decreased from 19.6% in 2013 to 10.7% in 2017. Compared to patients in the cluster with low-level SRNI over time, patients in the cluster with high-level persisting SRNI more often had a prior opportunistic infection of the central nervous system (CNS) (odds ratio [OR], 3.7; P < .001), imperfect adherence to antiretroviral therapy (ART) (OR, 2.8; P < .001), and depression (OR, 1.9; P < .001). CONCLUSIONS Although overall SRNI is decreasing in the SHCS, there is a group of patients with persisting SRNI over time. Past opportunistic infections of the CNS, imperfect adherence to ART, and depression were associated most with persisting SRNI. Patients with these characteristics should be preferentially tested for neurocognitive impairment.Although overall self-reported neurocognitive impairment (SRNI) is decreasing in the Swiss HIV Cohort Study, there is a group of patients with persisting SRNI over time, characterized by more past opportunistic infections of the central nervous system, imperfect adherence to antiretroviral therapy, and depression.
Collapse
Affiliation(s)
- Katharina Kusejko
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Luisa Salazar-Vizcaya
- Clinic for Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Dominique L Braun
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Philip E Tarr
- University Department of Medicine, Kantonsspital Baselland, University of Basel, Basel, Switzerland
| | - Enos Bernasconi
- Division of Infectious Diseases, Regional Hospital Lugano, Lugano, Switzerland
| | - Thanh Doco-Lecompte
- Department of Infectious Diseases, HIV Unit, Geneva University Hospital, University of Geneva, Geneva, Switzerland
| | - Matthias Cavassini
- Division of Infectious Diseases, Lausanne University Hospital, Lausanne, Switzerland
| | - Patrick Schmid
- Division of Infectious Diseases, Cantonal Hospital St Gallen, St. Gallen, Switzerland
| | - Renaud Du Pasquier
- Department of Clinical Neurosciences, Service of Neurology, Neuroimmunology Unit, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Christoph Hauser
- Clinic for Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Roger D Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
21
|
Brogan AJ, Talbird SE, Davis AE, La EM, Kumar PN. The Cost-Effectiveness and Budget Impact of Ibalizumab-uiyk for Adults with Multidrug-Resistant HIV-1 Infection in the United States. PHARMACOECONOMICS 2021; 39:421-432. [PMID: 33532919 DOI: 10.1007/s40273-020-00992-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Ibalizumab-uiyk (ibalizumab) is a first-in-class, long-acting, postattachment HIV-1 inhibitor for adults with multidrug-resistant (MDR) HIV-1 infection. This analysis examines the cost-effectiveness and budget impact of ibalizumab treatment for this difficult-to-treat population in the United States. METHODS A Markov model followed cohorts of adults with MDR HIV-1 infection through two final lines of antiretroviral therapy: ibalizumab + optimized background therapy (OBT) or OBT alone followed by nonsuppressive therapy. Model inputs were based on ibalizumab clinical trial data, market uptake projections, and published literature, with costs in 2019 dollars. The cost-effectiveness analysis assessed costs and health outcomes from a health care sector perspective for individuals receiving ibalizumab + OBT versus OBT alone over a lifetime time horizon. The budget-impact analysis estimated the impact on payer budgets of the introduction of ibalizumab over 3 years for a hypothetical commercial health plan. RESULTS Compared with individuals receiving OBT alone, individuals receiving ibalizumab + OBT incurred higher costs but lived longer, healthier lives, with an incremental cost of $133,040 per QALY gained. For a hypothetical commercial health plan with 1 million members, the introduction of ibalizumab + OBT was estimated to increase budgets by $217,260, $385,245, and $560,310 ($0.018, $0.032, and $0.047 per member per month) in years 1, 2, and 3, respectively. These results were found to be robust in sensitivity and scenario analyses. CONCLUSIONS Ibalizumab may represent a cost-effective and affordable option to improve health outcomes for individuals with MDR HIV-1 infection.
Collapse
Affiliation(s)
- Anita J Brogan
- RTI Health Solutions, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709, USA.
| | - Sandra E Talbird
- RTI Health Solutions, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Ashley E Davis
- RTI Health Solutions, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Elizabeth M La
- RTI Health Solutions, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709, USA
| | - Princy N Kumar
- Georgetown University School of Medicine, Georgetown University, Washington, DC, 20057, USA
| |
Collapse
|
22
|
Abela IA, Scherrer AU, Böni J, Yerly S, Klimkait T, Perreau M, Hirsch HH, Furrer H, Calmy A, Schmid P, Cavassini M, Bernasconi E, Günthard HF. Emergence of Drug Resistance in the Swiss HIV Cohort Study Under Potent Antiretroviral Therapy Is Observed in Socially Disadvantaged Patients. Clin Infect Dis 2021; 70:297-303. [PMID: 30843028 DOI: 10.1093/cid/ciz178] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/28/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The rate of acquired human immunodeficiency virus type 1 (HIV-1) drug resistance (ADR) has fallen dramatically since introduction of combined antiretroviral therapy (cART) in Switzerland. However, clinical experience indicates that there are still patients at risk of newly acquiring drug resistance despite having access to cART. Here, we characterized risk factors for ADR, to improve patient care and prevent emergence of drug resistance and treatment failure. METHODS We performed a case-control study to identify risk factors for ADR in all patients starting their first cART in the Swiss HIV Cohort Study (SHCS) since 1996. The SHCS is highly representative and includes >75% of patients receiving ART in Switzerland. To this end, we implemented a systematic medical chart review to obtain more detailed information on additional parameters, which are not routinely collected in the SHCS. The collected data were analyzed using univariable and multivariable conditional logistic regression. RESULTS We included in our study 115 cases and 115 matched controls. Unemployment (multivariable odds ratio [mOR], 2.9 [95% confidence interval {CI}, 1.3-6.4]; P = .008), African origin (mOR, 3.0 [95% CI, 1.0-9.2]; P = .047), comedication with anti-infectives (mOR, 3.7 [95% CI, 1.0-12.6]; P = .045), and symptoms of mental illness (mOR, 2.6 [95% CI, 1.2-5.5]; P = .012) were associated with ADR in the multivariable model. CONCLUSIONS Although ADR has become very rare with cART due to new potent therapies, patients in socially challenging life situations or presenting with mental health issues are at higher risk for drug resistance. Prompt identification and adequate support of these patients before ADR will prevent treatment failure and HIV-1 transmission.
Collapse
Affiliation(s)
- Irene A Abela
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich.,Institute of Medical Virology, University of Zurich
| | - Alexandra U Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich.,Institute of Medical Virology, University of Zurich
| | - Jürg Böni
- Institute of Medical Virology, University of Zurich
| | - Sabine Yerly
- Laboratory of Virology, Division of Infectious Diseases, Geneva University Hospitals
| | | | - Matthieu Perreau
- Division of Immunology and Allergy, University Hospital Lausanne
| | - Hans H Hirsch
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel
| | - Hansjakob Furrer
- Department of Infectious Diseases, Bern University Hospital, University of Bern
| | - Alexandra Calmy
- HIV/AIDS Unit, Infectious Disease Service, Geneva University Hospitals
| | - Patrick Schmid
- Division of Infectious Diseases, Cantonal Hospital St Gallen
| | | | - Enos Bernasconi
- Division of Infectious Diseases, Regional Hospital Lugano, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich.,Institute of Medical Virology, University of Zurich
| | | |
Collapse
|
23
|
Lombardi F, Giacomelli A, Armenia D, Lai A, Dusina A, Bezenchek A, Timelli L, Saladini F, Vichi F, Corsi P, Colao G, Bruzzone B, Gagliardini R, Callegaro A, Castagna A, Santoro MM. Prevalence and factors associated with HIV-1 multi-drug resistance over the past two decades in the Italian ARCA database. Int J Antimicrob Agents 2020; 57:106252. [PMID: 33259914 DOI: 10.1016/j.ijantimicag.2020.106252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 10/30/2020] [Accepted: 11/22/2020] [Indexed: 12/27/2022]
Abstract
Despite successful antiretroviral therapy (ART), patients infected with human immunodeficiency virus (HIV) can develop multi-class drug resistance (MDR). This retrospective study aimed to explore the prevalence of HIV-1 drug resistance over the past two decades by focusing on HIV-MDR and its predictors. ART-experienced patients with HIV with results from at least one plasma genotypic resistance test (GRT) from 1998 to 2018, from the Antiviral Response Cohort Analysis database, were included in this study. The temporal trend of resistance to any drug class was evaluated by considering all GRTs. Prevalence and predictors of HIV-MDR were analysed by consideration of cumulative GRTs. Among 15 628 isolates from 6802 patients, resistance to at least one drug class decreased sharply from 1998 to 2010 (1998-2001: 78%; 2008-2010: 59%; P<0.001) and then remained relatively constant at approximately 50% from 2011 to 2018, with the proportion of isolates with HIV-MDR also stable (approximately 9%). By evaluating factors associated with cumulative HIV-MDR, the following factors were found to be associated with increased risk of HIV-MDR on multi-variate analysis: male gender; sexual and vertical transmission; number of previous protease inhibitors, nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) and non-NRTIs; previous exposure to integrase strand transfer inhibitors, enfuvirtide and maraviroc; and co-infection with hepatitis B virus. In contrast, a nadir CD4 cell count ≥200 cells/mm3, starting first-line ART in 2008 or later and co-infection with hepatitis C virus were associated with lower risk of HIV-MDR. In conclusion, this study revealed that HIV-1 drug resistance has been stable since 2011 despite its dramatic decrease over the past two decades. HIV-MDR is still present, although at a lower rate, suggesting the need for continuous surveillance and accurate management of ART-experienced patients with HIV.
Collapse
Affiliation(s)
- Francesca Lombardi
- Università Cattolica del Sacro Cuore, Dipartimento di Sicurezza e Bioetica Sezione Malattie Infettive, Rome, Italy
| | - Andrea Giacomelli
- III Infectious Diseases Unit, ASST-FBF-Sacco, Milan, Italy; Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, Milan, Italy
| | - Daniele Armenia
- UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy; Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Alessia Lai
- Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, Milan, Italy
| | - Alex Dusina
- Università Cattolica del Sacro Cuore, Dipartimento di Sicurezza e Bioetica Sezione Malattie Infettive, Rome, Italy
| | | | | | - Francesco Saladini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Paola Corsi
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Grazia Colao
- Laboratory of Virology, Careggi Hospital, Florence, Italy
| | - Bianca Bruzzone
- Hygiene Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Roberta Gagliardini
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Rome, Italy
| | | | - Antonella Castagna
- Clinic of Infectious Diseases, Vita-Salute San Raffaele University, San Raffaele Scientific Institute, Milan, Italy
| | | | | |
Collapse
|
24
|
Substantial decline in heavily treated therapy-experienced persons with HIV with limited antiretroviral treatment options. AIDS 2020; 34:2051-2059. [PMID: 33055569 DOI: 10.1097/qad.0000000000002679] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Historically, a high burden of resistance to antiretroviral therapy (ART) in heavily treatment-experienced (HTE) persons with HIV (PWH) resulted in limited treatment options (LTOs). We evaluated the prevalence, risk factors, and virologic control of HTE PWH with LTO throughout the modern ART era. DESIGN We examined all ART-experienced PWH in care between 2000 and 2017 in the Centers for AIDS Research Network of Integrated Clinical Systems cohort. METHODS We computed the annual prevalence of HTE PWH with LTO defined as having two or less available classes with two or less active drugs per class based on genotypic data and cumulative antiretroviral resistance. We used multivariable Cox proportional hazards models to examine risk of LTO by 3-year study entry periods adjusting for demographic and clinical characteristics. RESULTS Among 27 133 ART-experienced PWH, 916 were classified as having LTO. The prevalence of PWH with LTO was 5.2-7.5% in 2000-2006, decreased to 1.8% in 2007, and remained less than 1% after 2012. Persons entering the study in 2009-2011 had an 80% lower risk of LTO compared with those entering in 2006-2008 (adjusted hazard ratio 0.20; 95% confidence interval: 0.09-0.42). We found a significant increase in undetectable HIV viral loads among PWH ever classified as having LTO from less than 30% in 2001 to more than 80% in 2011, comparable with persons who never had LTO. CONCLUSION Results of this large multicenter study show a dramatic decline in the prevalence of PWH with LTO to less than 1% with the availability of more potent drugs and a marked increase in virologic suppression in the current ART era.
Collapse
|
25
|
Pingarilho M, Pimentel V, Diogo I, Fernandes S, Miranda M, Pineda-Pena A, Libin P, Theys K, O. Martins MR, Vandamme AM, Camacho R, Gomes P, Abecasis A, on behalf of the Portuguese HIV-1 Resistance Study Group. Increasing Prevalence of HIV-1 Transmitted Drug Resistance in Portugal: Implications for First Line Treatment Recommendations. Viruses 2020; 12:E1238. [PMID: 33143301 PMCID: PMC7693025 DOI: 10.3390/v12111238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Treatment for All recommendations have allowed access to antiretroviral (ARV) treatment for an increasing number of patients. This minimizes the transmission of infection but can potentiate the risk of transmitted (TDR) and acquired drug resistance (ADR). OBJECTIVE To study the trends of TDR and ADR in patients followed up in Portuguese hospitals between 2001 and 2017. METHODS In total, 11,911 patients of the Portuguese REGA database were included. TDR was defined as the presence of one or more surveillance drug resistance mutation according to the WHO surveillance list. Genotypic resistance to ARV was evaluated with Stanford HIVdb v7.0. Patterns of TDR, ADR and the prevalence of mutations over time were analyzed using logistic regression. RESULTS AND DISCUSSION The prevalence of TDR increased from 7.9% in 2003 to 13.1% in 2017 (p < 0.001). This was due to a significant increase in both resistance to nucleotide reverse transcriptase inhibitors (NRTIs) and non-nucleotide reverse transcriptase inhibitors (NNRTIs), from 5.6% to 6.7% (p = 0.002) and 2.9% to 8.9% (p < 0.001), respectively. TDR was associated with infection with subtype B, and with lower viral load levels (p < 0.05). The prevalence of ADR declined from 86.6% in 2001 to 51.0% in 2017 (p < 0.001), caused by decreasing drug resistance to all antiretroviral (ARV) classes (p < 0.001). CONCLUSIONS While ADR has been decreasing since 2001, TDR has been increasing, reaching a value of 13.1% by the end of 2017. It is urgently necessary to develop public health programs to monitor the levels and patterns of TDR in newly diagnosed patients.
Collapse
Affiliation(s)
- Marta Pingarilho
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349–028 Lisbon, Portugal; (V.P.); (M.M.); (A.P.-P.); (M.R.O.M.); (A.-M.V.); (A.A.)
| | - Victor Pimentel
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349–028 Lisbon, Portugal; (V.P.); (M.M.); (A.P.-P.); (M.R.O.M.); (A.-M.V.); (A.A.)
| | - Isabel Diogo
- Laboratório de Biologia Molecular (LMCBM, SPC, CHLO-HEM), 1349-019 Lisbon, Portugal; (I.D.); (S.F.); (P.G.)
| | - Sandra Fernandes
- Laboratório de Biologia Molecular (LMCBM, SPC, CHLO-HEM), 1349-019 Lisbon, Portugal; (I.D.); (S.F.); (P.G.)
| | - Mafalda Miranda
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349–028 Lisbon, Portugal; (V.P.); (M.M.); (A.P.-P.); (M.R.O.M.); (A.-M.V.); (A.A.)
| | - Andrea Pineda-Pena
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349–028 Lisbon, Portugal; (V.P.); (M.M.); (A.P.-P.); (M.R.O.M.); (A.-M.V.); (A.A.)
| | - Pieter Libin
- Department of Microbiology and Immunology, KU Leuven, Clinical and Epidemiological Virology, Rega Institute for Medical Research, 3000 Leuven, Belgium; (P.L.); (K.T.); (R.C.)
- Artificial Intelligence Lab, Department of computer science, Vrije Universiteit Brussel, 1000 Brussels, Belgium
- Interuniversity Institute of Biostatistics and statistical Bioinformatics, Data Science Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Kristof Theys
- Department of Microbiology and Immunology, KU Leuven, Clinical and Epidemiological Virology, Rega Institute for Medical Research, 3000 Leuven, Belgium; (P.L.); (K.T.); (R.C.)
| | - M. Rosário O. Martins
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349–028 Lisbon, Portugal; (V.P.); (M.M.); (A.P.-P.); (M.R.O.M.); (A.-M.V.); (A.A.)
| | - Anne-Mieke Vandamme
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349–028 Lisbon, Portugal; (V.P.); (M.M.); (A.P.-P.); (M.R.O.M.); (A.-M.V.); (A.A.)
- Department of Microbiology and Immunology, KU Leuven, Clinical and Epidemiological Virology, Rega Institute for Medical Research, 3000 Leuven, Belgium; (P.L.); (K.T.); (R.C.)
| | - Ricardo Camacho
- Department of Microbiology and Immunology, KU Leuven, Clinical and Epidemiological Virology, Rega Institute for Medical Research, 3000 Leuven, Belgium; (P.L.); (K.T.); (R.C.)
| | - Perpétua Gomes
- Laboratório de Biologia Molecular (LMCBM, SPC, CHLO-HEM), 1349-019 Lisbon, Portugal; (I.D.); (S.F.); (P.G.)
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Superior de Ciências da Saúde Egas Moniz, 2829-511 Caparica, Portugal
| | - Ana Abecasis
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349–028 Lisbon, Portugal; (V.P.); (M.M.); (A.P.-P.); (M.R.O.M.); (A.-M.V.); (A.A.)
| | | |
Collapse
|
26
|
Lai A, Franzetti M, Bergna A, Saladini F, Bruzzone B, Di Giambenedetto S, Di Biagio A, Lo Caputo S, Santoro MM, Maggiolo F, Parisi SG, Rusconi S, Gianotti N, Balotta C. Marked decrease in acquired resistance to antiretrovirals in latest years in Italy. Clin Microbiol Infect 2020; 27:1038.e1-1038.e6. [PMID: 32979570 DOI: 10.1016/j.cmi.2020.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 08/25/2020] [Accepted: 09/15/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate acquired drug resistance in Italy in the 2009-2018 period. METHODS We analysed 3094 patients from the Italian ARCA database who had failed antiretroviral treatment and who had received a genotypic test after 6 months of treatment. Drug resistance mutations were identified using International AIDS Society (IAS)-USA tables and the Stanford HIVdb algorithm. The global burden of acquired resistance was calculated among all subjects with antiretroviral failure. Time trends and correlates of resistance were analysed using standard statistical tests. RESULTS Patients of non-European origin and non-B subtypes increased significantly from 11.5% (103/896) to 19.2% (33/172) and from 13.1% (141/1079) to 23.8% (53/223), respectively, over time. Overall, 14.5% (448/3094), 12.1% (374/3094) and 37.8% (1169/3094) of patients failed first, second and later lines, respectively. According to both IAS and HIVdb, in the study period resistance to any class, nucleoside reverse inhibitor, non-nucleoside reverse inhibitor, and protease inhibitors (PIs) declined significantly. Integrase strand transfer inhibitor (INSTI) resistance declined significantly from 31% (36/116) to 20.8% (41/197) according to HIVdb but not to IAS. Divergent data were highlighted regarding the proportion of non-European patients carrying any, PI and INSTI resistance using IAS tables compared with the Stanford HIVdb algorithm, as the former failed to detect a decrease in resistance while the latter indicates a reduction of 1.6-, 5- and 1.8-fold resistance for such drug classes. In the multivariate analysis, the risk of resistance increased in patients with a larger number of treatment lines and higher viraemia and decreased in those starting therapy in the last biennium of the study. DISCUSSION A marked reduction in drug resistance was observed over 10 years, compatible with higher genetic barrier and potency of new antiretrovirals. Nonetheless, concerns remain for subjects with non-B subtypes when using mutation lists instead of interpretation systems because of the extensive polymorphism of the protease region.
Collapse
Affiliation(s)
- Alessia Lai
- Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan, Italy.
| | - Marco Franzetti
- Infectious Diseases Unit, 'A. Manzoni' Hospital, Lecco, Italy
| | - Annalisa Bergna
- Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan, Italy
| | - Francesco Saladini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Bianca Bruzzone
- Hygiene Unit, Policlinico San Martino Hospital, Genoa, Italy
| | - Simona Di Giambenedetto
- Institute of Clinical Infectious Diseases, Catholic University of the Sacred Heart of Rome, Rome, Italy
| | - Antonio Di Biagio
- Infectious Diseases Clinic, Department of Health Sciences, University of Genoa, San Martino Hospital-IRCCS, Genoa, Italy
| | - Sergio Lo Caputo
- Clinic of Infectious Diseases, University Hospital Policlinico, University of Bari, Bari, Italy
| | | | - Franco Maggiolo
- Clinic of Infectious Diseases, Ospedali Riuniti, Bergamo, Italy
| | - Saverio G Parisi
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Stefano Rusconi
- Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan, Italy
| | | | - Claudia Balotta
- Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan, Italy
| |
Collapse
|
27
|
Omooja J, Nannyonjo M, Sanyu G, Nabirye SE, Nassolo F, Lunkuse S, Kapaata A, Segujja F, Kateete DP, Ssebaggala E, Bbosa N, Aling E, Nsubuga RN, Kaleebu P, Ssemwanga D. Rates of HIV-1 virological suppression and patterns of acquired drug resistance among fisherfolk on first-line antiretroviral therapy in Uganda. J Antimicrob Chemother 2020; 74:3021-3029. [PMID: 31257432 PMCID: PMC6753497 DOI: 10.1093/jac/dkz261] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 01/13/2023] Open
Abstract
Objectives We examined virological outcomes, patterns of acquired HIV drug resistance (ADR), correlates of virological failure (VF) and acquired drug resistance among fisherfolk on first-line ART. Methods We enrolled 1169 adults on ART for a median duration of 6, 12, 24, 36 and ≥48 months and used a pooled VL testing approach to identify VF (VL ≥1000 copies/mL). We performed genotyping among VF cases and determined correlates of VF and ADR by logistic regression. Results The overall virological suppression rate was 91.7% and ADR was detected in 71/97 (73.2%) VF cases. The most prevalent mutations were M184V/I (53.6%) for NRTIs and K103N (39.2%) for NNRTIs. Thymidine analogue mutations were detected in 21.6% of VF cases while PI mutations were absent. A zidovudine-based ART regimen, duration on ART (≥24 months) and secondary/higher education level were significantly associated with VF. A nevirapine-based regimen [adjusted OR (aOR): 1.87; 95% CI: 0.03–0.54)] and VL ≥10000 copies/mL (aOR: 3.48; 95% CI: 1.37–8.85) were ADR correlates. The pooling strategies for VL testing with a negative predictive value (NPV) of ≥95.2% saved US $20320 (43.5%) in VL testing costs. Conclusions We observed high virological suppression rates among these highly mobile fisherfolk; however, there was widespread ADR among those with VF at the first VL testing prior to intensive adherence counselling. Timely treatment switching and adherence support is recommended for better treatment outcomes. Adoption of pooled VL testing could be cost effective, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Jonah Omooja
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda.,Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Maria Nannyonjo
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Grace Sanyu
- Uganda Virus Research Institute, Entebbe, Uganda
| | - Stella E Nabirye
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Faridah Nassolo
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Sandra Lunkuse
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Anne Kapaata
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Farouk Segujja
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda.,Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - David Patrick Kateete
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda.,Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Eric Ssebaggala
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Nicholas Bbosa
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Emmanuel Aling
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Rebecca N Nsubuga
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Pontiano Kaleebu
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda.,Uganda Virus Research Institute, Entebbe, Uganda
| | - Deogratius Ssemwanga
- Medical Research Council (MRC)/Uganda Virus Research Institute (UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda.,Uganda Virus Research Institute, Entebbe, Uganda
| |
Collapse
|
28
|
Rhee SY, Clutter D, Hare CB, Tchakoute CT, Sainani K, Fessel WJ, Hurley L, Slome S, Pinsky BA, Silverberg MJ, Shafer RW. Virological Failure and Acquired Genotypic Resistance Associated With Contemporary Antiretroviral Treatment Regimens. Open Forum Infect Dis 2020; 7:ofaa316. [PMID: 32904894 PMCID: PMC7462367 DOI: 10.1093/ofid/ofaa316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022] Open
Abstract
Background There are few descriptions of virologic failure (VF) and acquired drug resistance (HIVDR) in large cohorts initiating contemporary antiretroviral therapy (ART). Methods We studied all persons with HIV (PWH) in a California clinic population initiating ART between 2010 and 2017. VF was defined as not attaining virologic suppression, discontinuing ART, or virologic rebound prompting change in ART. Results During the study, 2315 PWH began ART. Six companion drugs were used in 93.3% of regimens: efavirenz, elvitegravir/c, dolutegravir, darunavir/r, rilpivirine, and raltegravir. During a median follow-up of 36 months, 214 (9.2%) PWH experienced VF (2.8 per 100 person-years) and 62 (2.7%) experienced HIVDR (0.8 per 100 person-years). In multivariable analyses, younger age, lower CD4 count, higher virus load, and atazanavir/r were associated with increased VF risk; lower CD4 count, higher virus load, and nevirapine were associated with increased HIVDR risk. Compared with efavirenz, dolutegravir, raltegravir, and darunavir were associated with reduced HIVDR risk. Risks of VF and HIVDR were not significantly associated with ART initiation year. Of the 62 PWH with HIVDR, 42 received an non-nucleoside RT inhibitor (NNRTI), 15 an integrase-strand transfer inhibitor (INSTI), and 5 a protease inhibitor (PI). Among those with HIVDR on an NNRTI or first-generation INSTI, 59% acquired dual class resistance and 29% developed tenofovir resistance; those receiving a PI or dolutegravir developed just M184V. Conclusions Despite the frequent use of contemporary ART regimens, VF and HIVDR continue to occur. Further efforts are required to improve long-term ART virological responses to prevent the consequences of ongoing HIV-1 replication including virus transmission and HIVDR.
Collapse
Affiliation(s)
- Soo-Yon Rhee
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Dana Clutter
- Department of Infectious Diseases, Kaiser Permanente Northern California, South San Francisco, California, USA
| | - C Bradley Hare
- Department of Infectious Diseases, Kaiser Permanente Northern California, San Francisco, California, USA
| | - Christophe T Tchakoute
- Division of Epidemiology and Population Health, Department of Medicine, Stanford University, Stanford, California, USA
| | - Kristin Sainani
- Division of Epidemiology and Population Health, Department of Medicine, Stanford University, Stanford, California, USA
| | - W Jeffrey Fessel
- Department of Infectious Diseases, Kaiser Permanente Northern California, San Francisco, California, USA
| | - Leo Hurley
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Sally Slome
- Department of Infectious Diseases, Kaiser Permanente Northern California, Oakland, California, USA
| | - Benjamin A Pinsky
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Michael J Silverberg
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Robert W Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
29
|
Giacomelli A, Pezzati L, Rusconi S. The crosstalk between antiretrovirals pharmacology and HIV drug resistance. Expert Rev Clin Pharmacol 2020; 13:739-760. [PMID: 32538221 DOI: 10.1080/17512433.2020.1782737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The clinical development of antiretroviral drugs has been followed by a rapid and concomitant development of HIV drug resistance. The development and spread of HIV drug resistance is due on the one hand to the within-host intrinsic HIV evolutionary rate and on the other to the wide use of low genetic barrier antiretrovirals. AREAS COVERED We searched PubMed and Embase on 31 January 2020, for studies reporting antiretroviral resistance and pharmacology. In this review, we assessed the molecular target and mechanism of drug resistance development of the different antiretroviral classes focusing on the currently approved antiretroviral drugs. Then, we assessed the main pharmacokinetic/pharmacodynamic of the antiretrovirals. Finally, we retraced the history of antiretroviral treatment and its interconnection with antiretroviral worldwide resistance development both in , and middle-income countries in the perspective of 90-90-90 World Health Organization target. EXPERT OPINION Drug resistance development is an invariably evolutionary driven phenomenon, which challenge the 90-90-90 target. In high-income countries, the antiretroviral drug resistance seems to be stable since the last decade. On the contrary, multi-intervention strategies comprehensive of broad availability of high genetic barrier regimens should be implemented in resource-limited setting to curb the rise of drug resistance.
Collapse
Affiliation(s)
- Andrea Giacomelli
- III Infectious Disease Unit, ASST-FBF-Sacco , Milan, Italy.,Department of Biomedical and Clinical Sciences DIBIC L. Sacco, University of Milan , Milan, Italy
| | - Laura Pezzati
- III Infectious Disease Unit, ASST-FBF-Sacco , Milan, Italy.,Department of Biomedical and Clinical Sciences DIBIC L. Sacco, University of Milan , Milan, Italy
| | - Stefano Rusconi
- III Infectious Disease Unit, ASST-FBF-Sacco , Milan, Italy.,Department of Biomedical and Clinical Sciences DIBIC L. Sacco, University of Milan , Milan, Italy
| |
Collapse
|
30
|
Günthard HF, Calvez V, Paredes R, Pillay D, Shafer RW, Wensing AM, Jacobsen DM, Richman DD. Human Immunodeficiency Virus Drug Resistance: 2018 Recommendations of the International Antiviral Society-USA Panel. Clin Infect Dis 2020; 68:177-187. [PMID: 30052811 PMCID: PMC6321850 DOI: 10.1093/cid/ciy463] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/28/2018] [Indexed: 12/16/2022] Open
Abstract
Background Contemporary antiretroviral therapies (ART) and management strategies have diminished both human immunodeficiency virus (HIV) treatment failure and the acquired resistance to drugs in resource-rich regions, but transmission of drug-resistant viruses has not similarly decreased. In low- and middle-income regions, ART roll-out has improved outcomes, but has resulted in increasing acquired and transmitted resistances. Our objective was to review resistance to ART drugs and methods to detect it, and to provide updated recommendations for testing and monitoring for drug resistance in HIV-infected individuals. Methods A volunteer panel of experts appointed by the International Antiviral (formerly AIDS) Society–USA reviewed relevant peer-reviewed data that were published or presented at scientific conferences. Recommendations were rated according to the strength of the recommendation and quality of the evidence, and reached by full panel consensus. Results Resistance testing remains a cornerstone of ART. It is recommended in newly-diagnosed individuals and in patients in whom ART has failed. Testing for transmitted integrase strand-transfer inhibitor resistance is currently not recommended, but this may change as more resistance emerges with widespread use. Sanger-based and next-generation sequencing approaches are each suited for genotypic testing. Testing for minority variants harboring drug resistance may only be considered if treatments depend on a first-generation nonnucleoside analogue reverse transcriptase inhibitor. Different HIV-1 subtypes do not need special considerations regarding resistance testing. Conclusions Testing for HIV drug resistance in drug-naive individuals and in patients in whom antiretroviral drugs are failing, and the appreciation of the role of testing, are crucial to the prevention and management of failure of ART.
Collapse
Affiliation(s)
- Huldrych F Günthard
- University Hospital Zürich and Institute of Medical Virology, University of Zurich, Switzerland
| | - Vincent Calvez
- Pierre et Marie Curie University and Pitié-Salpêtriere Hospital, Paris, France
| | - Roger Paredes
- Infectious Diseases Service and IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Africa Health Research Institute, KwaZulu Natal, South Africa
| | | | | | | | | | - Douglas D Richman
- Veterans Affairs San Diego Healthcare System and University of California San Diego
| |
Collapse
|
31
|
Abstract
OBJECTIVE To analyze HIV drug resistance among MSM recruited for participation in the HPTN 078 study, which evaluated methods for achieving and maintaining viral suppression in HIV-infected MSM. METHODS Individuals were recruited at four study sites in the United States (Atlanta, Georgia; Baltimore, Maryland; Birmingham, Alabama; and Boston, Massachusetts; 2016-2017). HIV genotyping was performed using samples collected at study screening or enrollment. HIV drug resistance was evaluated using the Stanford v8.7 algorithm. A multiassay algorithm was used to identify individuals with recent HIV infection. Clustering of HIV sequences was evaluated using phylogenetic methods. RESULTS High-level HIV drug resistance was detected in 44 (31%) of 142 individuals (Atlanta: 21%, Baltimore: 29%, Birmingham: 53%, Boston: 26%); 12% had multiclass resistance, 16% had resistance to tenofovir or emtricitabine, and 8% had resistance to integrase strand transfer inhibitors (INSTIs); 3% had intermediate-level resistance to second-generation INSTIs. In a multivariate model, self-report of ever having been on antiretroviral therapy (ART) was associated with resistance (P = 0.005). One of six recently infected individuals had drug resistance. Phylogenetic analysis identified five clusters of study sequences; two clusters had shared resistance mutations. CONCLUSION High prevalence of drug resistance was observed among MSM. Some had multiclass resistance, resistance to drugs used for preexposure prophylaxis (PrEP), and INSTI resistance. These findings highlight the need for improved HIV care in this high-risk population, identification of alternative regimens for PrEP, and inclusion of integrase resistance testing when selecting ART regimens for MSM in the United States.
Collapse
|
32
|
Lodi S, Günthard HF, Gill J, Phillips AN, Dunn D, Vu Q, Siemieniuk R, Garcia F, Logan R, Jose S, Bucher HC, Scherrer AU, Reiss P, van Sighem A, Boender TS, Porter K, Gilson R, Paraskevis D, Simeon M, Vourli G, Moreno S, Jarrin I, Sabin C, Hernán MA. Effectiveness of Transmitted Drug Resistance Testing Before Initiation of Antiretroviral Therapy in HIV-Positive Individuals. J Acquir Immune Defic Syndr 2019; 82:314-320. [PMID: 31609929 PMCID: PMC7830777 DOI: 10.1097/qai.0000000000002135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND For people living with HIV, major guidelines in high-income countries recommend testing for transmitted drug resistance (TDR) to guide the choice of first-line antiretroviral therapy (ART). However, individuals who fail a first-line regimen can now be switched to one of several effective regimens. Therefore, the virological and clinical benefit of TDR testing needs to be evaluated. METHODS We included individuals from the HIV-CAUSAL Collaboration who enrolled <6 months of HIV diagnosis between 2006 and 2015, were ART-naive, and had measured CD4 count and HIV-RNA. Follow-up started at the date when all inclusion criteria were first met (baseline). We compared 2 strategies: (1) TDR testing within 3 months of baseline versus (2) no TDR testing. We used inverse probability weighting to estimate the 5-year proportion and hazard ratios (HRs) of virological suppression (confirmed HIV-RNA <50 copies/mL), and of AIDS or death under both strategies. RESULTS Of 25,672 eligible individuals (82% males, 52% diagnosed in 2010 or later), 17,189 (67%) were tested for TDR within 3 months of baseline. Of these, 6% had intermediate- or high-level TDR to any antiretroviral drug. The estimated 5-year proportion virologically suppressed was 77% under TDR testing and 74% under no TDR testing; HR 1.06 (95% confidence interval: 1.03 to 1.19). The estimated 5-year risk of AIDS or death was 6% under both strategies; HR 1.03 (95% confidence interval: 0.95 to 1.12). CONCLUSIONS TDR prevalence was low. Although TDR testing improved virological response, we found no evidence that it reduced the incidence of AIDS or death in first 5 years after diagnosis.
Collapse
Affiliation(s)
- Sara Lodi
- Boston University School of Public Health, Boston, MA
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Zürich, Switzerland
| | - John Gill
- University of Calgary, Calgary, Alberta, Canada
- Southern Alberta Clinic, Calgary, Alberta, Canada
| | - Andrew N Phillips
- Institute for Global Health, University College London, London, United Kingdom
| | - David Dunn
- Institute for Global Health, University College London, London, United Kingdom
| | - Quang Vu
- University of Calgary, Calgary, Alberta, Canada
| | - Reed Siemieniuk
- Southern Alberta Clinic, Calgary, Alberta, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | | | - Roger Logan
- Harvard T.H. Chan School of Public Health, Boston, MA
| | - Sophie Jose
- Institute for Global Health, University College London, London, United Kingdom
| | - Heiner C Bucher
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandra U Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Zürich, Switzerland
| | - Peter Reiss
- Stichting HIV Monitoring, Amsterdam, the Netherlands
- Division of Infectious Diseases, Department of Global Health, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | | | | | - Kholoud Porter
- Institute for Global Health, University College London, London, United Kingdom
| | - Richard Gilson
- Institute for Global Health, University College London, London, United Kingdom
| | | | | | - Georgia Vourli
- National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Santiago Moreno
- Ramón y Cajal Hospital, IRYCIS, Madrid, Spain
- University of Alcalá de Henares, Madrid, Spain
| | - Inmaculada Jarrin
- Centro Nacional de Epidemiologia, Instituto de Salud Carlos III, Madrid, Spain
| | - Caroline Sabin
- Institute for Global Health, University College London, London, United Kingdom
| | - Miguel A Hernán
- Harvard T.H. Chan School of Public Health, Boston, MA
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA
| |
Collapse
|
33
|
Mbunkah HA, Marzel A, Schmutz S, Kok YL, Zagordi O, Shilaih M, Nsanwe NN, Mbu ET, Besong LM, Sama BA, Orock E, Kouyos RD, Günthard HF, Metzner KJ. Low prevalence of transmitted HIV-1 drug resistance detected by a dried blood spot (DBS)-based next-generation sequencing (NGS) method in newly diagnosed individuals in Cameroon in the years 2015-16. J Antimicrob Chemother 2019; 73:1917-1929. [PMID: 29635462 DOI: 10.1093/jac/dky103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/02/2018] [Indexed: 11/13/2022] Open
Abstract
Objectives To determine the most recent prevalence, transmission patterns and risk factors of transmitted drug-resistance mutations (TDRMs) in Cameroon, we initiated a multicentre study monitoring HIV-1 drug resistance in newly HIV-1-diagnosed individuals using a novel next-generation sequencing (NGS) assay applicable to fingerprick dried blood spot (DBS) samples. Methods Fingerprick DBS samples and questionnaires were collected from 360 newly HIV-1-diagnosed individuals in four hospitals in urban areas in Cameroon in the years 2015-16. We developed an HIV-1 protease and reverse transcriptase drug resistance genotyping assay applicable to DBS samples and HIV-1 genomes of groups M, N and O. The WHO 2009 list of mutations for surveillance of transmitted drug-resistant HIV strains was used to analyse TDRMs. Results Applying our 'DBS-NGS-genotypic resistance test', baseline HIV-1 drug resistance data were successfully obtained from 82.8% (298/360) of newly diagnosed individuals. At nucleotide frequencies >15%, TDRMs to NRTIs were observed in 3.0% (9/298), to NNRTIs in 4.0% (12/298) and to PIs in 1.3% (3/240). The NNRTI mutation K103N was most commonly detected (2.7%). Expanding the analysis to low-abundance TDRMs, i.e. 3%-15%, 12 additional individuals (4.0%) harbouring TDRMs were identified. Having unprotected sex with a known HIV-1-positive person was significantly associated with the transmission of DRMs (adjusted OR 9.6; 95% CI 1.79-51.3). Conclusions The prevalence of transmitted HIV-1 drug resistance is currently low in the study sites in Cameroon. Evidence of some risky sexual behaviours depicts a public health problem with possible implications for the prevention of new HIV-1 infections.
Collapse
Affiliation(s)
- Herbert A Mbunkah
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.,Life Science Zurich Graduate School, Microbiology and Immunology PhD Programme, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Alex Marzel
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Stefan Schmutz
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Yik Lim Kok
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Osvaldo Zagordi
- Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Mohaned Shilaih
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland
| | - Ndi N Nsanwe
- Regional Hospital Bamenda, PO Box 863, Mankon-Bamenda, Cameroon
| | - Eyongetah T Mbu
- Regional Hospital Bamenda, PO Box 863, Mankon-Bamenda, Cameroon
| | - Lydia M Besong
- District Hospital Kumba, Meme Division, South-West Region, Cameroon
| | - Bella A Sama
- District Hospital Ndop, Ngoketunjia Division, North-West Region, Cameroon
| | - Emmanuel Orock
- Regional Hospital Ngaoundere, Avenue Rue Ahidjo Ngaoundéré, Adamawa, Cameroon
| | - Roger D Kouyos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Karin J Metzner
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Institute of Medical Virology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| |
Collapse
|
34
|
Stirrup OT, Sabin CA, Phillips AN, Williams I, Churchill D, Tostevin A, Hill T, Dunn DT, Asboe D, Pozniak A, Cane P, Chadwick D, Churchill D, Clark D, Collins S, Delpech V, Douthwaite S, Dunn D, Fearnhill E, Porter K, Tostevin A, Stirrup O, Fraser C, Geretti AM, Gunson R, Hale A, Hué S, Lazarus L, Leigh-Brown A, Mbisa T, Mackie N, Orkin C, Nastouli E, Pillay D, Phillips A, Sabin C, Smit E, Templeton K, Tilston P, Volz E, Williams I, Zhang H, Fairbrother K, Dawkins J, O’Shea S, Mullen J, Cox A, Tandy R, Fawcett T, Hopkins M, Booth C, Renwick L, Renwick L, Schmid ML, Payne B, Hubb J, Dustan S, Kirk S, Bradley-Stewart A, Hill T, Jose S, Thornton A, Huntington S, Glabay A, Shidfar S, Lynch J, Hand J, de Souza C, Perry N, Tilbury S, Youssef E, Gazzard B, Nelson M, Mabika T, Mandalia S, Anderson J, Munshi S, Post F, Adefisan A, Taylor C, Gleisner Z, Ibrahim F, Campbell L, Baillie K, Gilson R, Brima N, Ainsworth J, Schwenk A, Miller S, Wood C, Johnson M, Youle M, Lampe F, Smith C, Tsintas R, Chaloner C, Hutchinson S, Walsh J, Mackie N, Winston A, et alStirrup OT, Sabin CA, Phillips AN, Williams I, Churchill D, Tostevin A, Hill T, Dunn DT, Asboe D, Pozniak A, Cane P, Chadwick D, Churchill D, Clark D, Collins S, Delpech V, Douthwaite S, Dunn D, Fearnhill E, Porter K, Tostevin A, Stirrup O, Fraser C, Geretti AM, Gunson R, Hale A, Hué S, Lazarus L, Leigh-Brown A, Mbisa T, Mackie N, Orkin C, Nastouli E, Pillay D, Phillips A, Sabin C, Smit E, Templeton K, Tilston P, Volz E, Williams I, Zhang H, Fairbrother K, Dawkins J, O’Shea S, Mullen J, Cox A, Tandy R, Fawcett T, Hopkins M, Booth C, Renwick L, Renwick L, Schmid ML, Payne B, Hubb J, Dustan S, Kirk S, Bradley-Stewart A, Hill T, Jose S, Thornton A, Huntington S, Glabay A, Shidfar S, Lynch J, Hand J, de Souza C, Perry N, Tilbury S, Youssef E, Gazzard B, Nelson M, Mabika T, Mandalia S, Anderson J, Munshi S, Post F, Adefisan A, Taylor C, Gleisner Z, Ibrahim F, Campbell L, Baillie K, Gilson R, Brima N, Ainsworth J, Schwenk A, Miller S, Wood C, Johnson M, Youle M, Lampe F, Smith C, Tsintas R, Chaloner C, Hutchinson S, Walsh J, Mackie N, Winston A, Weber J, Ramzan F, Carder M, Leen C, Wilson A, Morris S, Gompels M, Allan S, Palfreeman A, Lewszuk A, Kegg S, Faleye A, Ogunbiyi V, Mitchell S, Hay P, Kemble C, Martin F, Russell-Sharpe S, Gravely J, Allan S, Harte A, Tariq A, Spencer H, Jones R, Pritchard J, Cumming S, Atkinson C, Mital D, Edgell V, Allen J, Ustianowski A, Murphy C, Gunder I, Trevelion R, Babiker A. Associations between baseline characteristics, CD4 cell count response and virological failure on first-line efavirenz + tenofovir + emtricitabine for HIV. J Virus Erad 2019. [DOI: 10.1016/s2055-6640(20)30037-6] [Show More Authors] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
35
|
New antiretroviral agent use affects prevalence of HIV drug resistance in clinical care populations. AIDS 2018; 32:2593-2603. [PMID: 30134298 DOI: 10.1097/qad.0000000000001990] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To estimate the prevalence of HIV drug resistance over time and identify risk factors for multiclass resistance. DESIGN Prospective clinical cohort of HIV-infected patients at the University of North Carolina. METHODS Among antiretroviral therapy (ART)-experienced patients in care 2000-2016, we estimated annual prevalences of cumulative resistance, defined as at least one major mutation by drug class. Clinical data and multiple imputation were used when genotypic data were missing, and mutations were carried forward in time. We estimated resistance odds ratios comparing characteristics of patients in care in 2016. RESULTS A total of 3682 patients contributed 23 169 person-years. Prevalence of at least one major resistance mutation, irrespective of viral suppression, peaked in 2005 with 49% (95% confidence interval 46, 52) and decreased to 38% (35, 40) in 2016. Resistance to nucleoside reverse transcriptase inhibitors, protease inhibitors, and nonnucleoside reverse transcriptase inhibitors also peaked in 2005-2007 and decreased to 28 (26, 31), 14 (12, 16), and 27% (24, 29) in 2016, respectively. In 2016, prevalence of integrase strand transfer inhibitor (INSTI) resistance was 2% (1, 3) and triple-class resistance 10% (9, 12). Over the study period, cumulative resistance was frequent among patients with detectable viremia, but uncommon among patients initiating ART post-2007. Among 1553 patients in care in 2016, ART initiation at an older age, with an INSTI, and with higher CD4 cell counts were associated with resistance to fewer or no classes. CONCLUSION Prevalence of resistance to older ART classes has decreased in the last 10 years in this clinical cohort, whereas INSTI resistance has increased but remained very low. Patients with viremia continue to have a high burden of resistance even if they initiated ART recently.
Collapse
|
36
|
Karkashadze E, Dvali N, Bolokadze N, Sharvadze L, Gabunia P, Karchava M, Tchelidze T, Tsertsvadze T, DeHovitz J, Del Rio C, Chkhartishvili N. Epidemiology of human immunodeficiency virus (HIV) drug resistance in HIV patients with virologic failure of first-line therapy in the country of Georgia. J Med Virol 2018; 91:235-240. [PMID: 29905958 DOI: 10.1002/jmv.25245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/05/2018] [Indexed: 11/08/2022]
Abstract
Human immunodeficiency virus (HIV) drug resistance is a major threat to the sustained impact of antiretroviral therapy (ART). We studied the epidemiology of drug resistance in the country of Georgia. The study included all adult patients who experienced virologic failure on first line ART and received HIV drug resistance testing between 2005 and 2016. The Stanford HIV Sequence Database was used for interpretation of the resistance data. Patient-level data were extracted from the national AIDS health information system. Of the 447 patients included, 85.5% harbored the subtype A6 virus, 8.0% - subtype B, 2.9% - subtype G, and other subtypes were <1%. The most frequent first-line regimens were Tenofovir/Emtricitabine/Efavirenz (28.4%), Zidovudine/Lamivudine/Efavirenz (28.4%), and Abacavir/Lamivudine/Efavirenz (15.9%). A total of 85.0% of the patients with treatment failure developed at least one drug resistance mutation affecting their susceptibility to ART. The most frequent nucleoside reverse transcriptase inhibitor mutations were M184V (65.3%), K65R (19.7%) and L74V (17.0%). At least three thymidine analogue mutations were detected in 6.3% of the patients. From non-nucleoside reverse transcriptase inhibitor mutations, G190S was shown to be the most prevalent (49.4%), followed by K101E (27.10%) and K103N (24.4%). G190S and K101E were more common in subtype A as compared with non-A viruses (G190S: 54.9% vs 11.3%, P < 0.0001; K101E: 29.8% vs 11.3%, P = 0.005). On the other hand, K103N was more frequent in non-A subtypes (43.4%) compared with subtype A (22.2%), P = 0.0008. A majority of persons failing on ART had HIV drug resistance. Drug resistance patterns may vary by subtype. K65R mutation remains below 20%, but given the high use of Tenofovir in the country, continuing surveillance of drug resistance is needed.
Collapse
Affiliation(s)
| | - Natia Dvali
- Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia
| | - Natalia Bolokadze
- Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia
| | - Lali Sharvadze
- Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia.,Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Pati Gabunia
- Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia
| | - Marine Karchava
- Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia
| | - Tamar Tchelidze
- Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia
| | - Tengiz Tsertsvadze
- Infectious Diseases, AIDS and Clinical Immunology Research Center, Tbilisi, Georgia.,Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | | | | | | |
Collapse
|
37
|
Stirrup OT, Dunn DT, Tostevin A, Sabin CA, Pozniak A, Asboe D, Cox A, Orkin C, Martin F, Cane P. Risk factors and outcomes for the Q151M and T69 insertion HIV-1 resistance mutations in historic UK data. AIDS Res Ther 2018; 15:11. [PMID: 29661246 PMCID: PMC5902836 DOI: 10.1186/s12981-018-0198-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/28/2018] [Indexed: 01/24/2023] Open
Abstract
Background The prevalence of HIV-1 resistance to antiretroviral therapies (ART) has declined in high-income countries over recent years, but drug resistance remains a substantial concern in many low and middle-income countries. The Q151M and T69 insertion (T69i) resistance mutations in the viral reverse transcriptase gene can reduce susceptibility to all nucleoside/tide analogue reverse transcriptase inhibitors, motivating the present study to investigate the risk factors and outcomes associated with these mutations. Methods We considered all data in the UK HIV Drug Resistance Database for blood samples obtained in the period 1997–2014. Where available, treatment history and patient outcomes were obtained through linkage to the UK Collaborative HIV Cohort study. A matched case–control approach was used to assess risk factors associated with the appearance of each of the mutations in ART-experienced patients, and survival analysis was used to investigate factors associated with viral suppression. A further analysis using matched controls was performed to investigate the impact of each mutation on survival. Results A total of 180 patients with Q151M mutation and 85 with T69i mutation were identified, almost entirely from before 2006. Occurrence of both the Q151M and T69i mutations was strongly associated with cumulative period of virological failure while on ART, and for Q151M there was a particular positive association with use of stavudine and negative association with use of boosted-protease inhibitors. Subsequent viral suppression was negatively associated with viral load at sequencing for both mutations, and for Q151M we found a negative association with didanosine use but a positive association with boosted-protease inhibitor use. The results obtained in these analyses were also consistent with potentially large associations with other drugs. Analyses were inconclusive regarding associations between the mutations and mortality, but mortality was high for patients with low CD4 at detection. Conclusions The Q151M and T69i resistance mutations are now very rare in the UK. Our results suggest that good outcomes are possible for people with these mutations. However, in this historic sample, viral load and CD4 at detection were important factors in determining prognosis. Electronic supplementary material The online version of this article (10.1186/s12981-018-0198-7) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Lodi S, Günthard HF, Dunn D, Garcia F, Logan R, Jose S, Bucher HC, Scherrer AU, Schneider MP, Egger M, Glass TR, Reiss P, van Sighem A, Boender TS, Phillips AN, Porter K, Hawkins D, Moreno S, Monge S, Paraskevis D, Simeon M, Vourli G, Sabin C, Hernán MA. Effect of immediate initiation of antiretroviral treatment on the risk of acquired HIV drug resistance. AIDS 2018; 32:327-335. [PMID: 29135583 PMCID: PMC5758415 DOI: 10.1097/qad.0000000000001692] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We estimated and compared the risk of clinically identified acquired drug resistance under immediate initiation [the currently recommended antiretroviral therapy (ART) initiation strategy], initiation with CD4 cell count less than 500 cells/μl and initiation with CD4 cell count less than 350 cells/μl. DESIGN Cohort study based on routinely collected data from the HIV-CAUSAL collaboration. METHODS For each individual, baseline was the earliest time when all eligibility criteria (ART-naive, AIDS free, and others) were met after 1999. Acquired drug resistance was defined using the Stanford classification as resistance to any antiretroviral drug that was clinically identified at least 6 months after ART initiation. We used the parametric g-formula to adjust for time-varying (CD4 cell count, HIV RNA, AIDS, ART regimen, and drug resistance testing) and baseline (calendar period, mode of acquisition, sex, age, geographical origin, ethnicity and cohort) characteristics. RESULTS In 50 981 eligible individuals, 10% had CD4 cell count more than 500 cells/μl at baseline, and 63% initiated ART during follow-up. Of 2672 tests for acquired drug resistance, 794 found resistance. The estimated 7-year risk (95% confidence interval) of acquired drug resistance was 3.2% (2.8,3.5) for immediate initiation, 3.1% (2.7,3.3) for initiation with CD4 cell count less than 500 cells/μl, and 2.8% (2.5,3.0) for initiation with CD4 cell count less than 350 cells/μl. In analyses restricted to individuals with baseline in 2005-2015, the corresponding estimates were 1.9% (1.8, 2.5), 1.9% (1.7, 2.4), and 1.8% (1.7, 2.2). CONCLUSION Our findings suggest that the risk of acquired drug resistance is very low, especially in recent calendar periods, and that immediate ART initiation only slightly increases the risk. It is unlikely that drug resistance will jeopardize the proven benefits of immediate ART initiation.
Collapse
Affiliation(s)
- Sara Lodi
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | | | | | - Roger Logan
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Heiner C Bucher
- Basel Institute for Clinical Epidemiology and Biostatistics, University Hospital Basel, University of Basel, Basel
| | - Alexandra U Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Marie-Paule Schneider
- Department of Ambulatory Care and Community Medicine, University of Lausanne, Lausanne
- Community Pharmacy, School of Pharmaceutical Sciences, University of Geneva, Geneva
| | - Matthias Egger
- Institute of Social and Preventive Medicine, University of Bern, Bern
| | - Tracy R Glass
- Swiss Tropical and Public Health Institute
- University of Basel, Basel, Switzerland
| | - Peter Reiss
- Stichting HIV Monitoring
- Division of Infectious Diseases, Department of Global Health, Academic Medical Centre, University of Amsterdam
- Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| | | | | | | | | | | | - Santiago Moreno
- IRYCIS, Ramón y Cajal Hospital
- University of Alcalá de Henares
| | - Susana Monge
- University of Alcalá de Henares
- National Centre of Epidemiology - ISCIII, Madrid, Spain
| | | | | | - Georgia Vourli
- National and Kapodistrian University of Athens Medical School, Athens
| | | | - Miguel A Hernán
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard-MIT Division of Health Sciences and Technology, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Abstract
BACKGROUND The rate of transmitted drug resistance (TDR) may increase with wider use of antiretroviral therapy and can contribute to therapeutic failure. We analysed time trends in TDR among HIV seroconverters. METHODS Using CASCADE data of individuals with well estimated dates of HIV seroconversion, we examined HIV nucleotide sequences collected prior to antiretroviral therapy use from 1996-2012. All samples were taken within 12 months of testing HIV positive. Using logistic regression, we examined the association between TDR and year of seroconversion, adjusting for confounders. RESULTS Of 4717 individuals seroconverting between 1996 and 2012, median (IQR) age at seroconversion was 33 (27, 39) years. The majority (3839; 92%) were male, mainly exposed through MSM (3767; 80%), and infected with subtype B (3464; 73%). Overall, 515 (11%) individuals had at least one drug resistance-related mutation; 280 individuals with nucleoside reverse transcriptase, 185 with nonnucleoside reverse transcriptase, and 144 with protease inhibitor mutations. Estimated TDR prevalence was 19.4% (8.2, 36.0) in 1996, significantly decreasing to 8.5% (5.9, 11.9) in 2012 [odds ratio (OR; 95% confidence interval (CI)) = 0.92 (0.90, 0.95) per year increase]. Individuals exposed through sex between men and women were significantly less likely to have been infected with a drug-resistant strain [OR (95% CI) = 0.59 (0.41, 0.87) compared with MSM], and there was marginal evidence that sampling during acute infection was associated with higher odds of resistance [OR (95% CI) = 1.20 (0.97, 1.7), P = 0.093] compared with later sampling. CONCLUSION TDR has decreased over calendar time although a significant proportion of new infections still carry resistance-related mutations.
Collapse
|
40
|
Zhukova A, Cutino-Moguel T, Gascuel O, Pillay D. The Role of Phylogenetics as a Tool to Predict the Spread of Resistance. J Infect Dis 2017; 216:S820-S823. [PMID: 29029155 DOI: 10.1093/infdis/jix411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Drug resistance mutations emerge in genetic sequences of HIV through drug-selective pressure. Drug resistance can be transmitted. In this review we discuss phylogenetic methods used to study the emergence of drug resistance and the spread of resistant viruses.
Collapse
Affiliation(s)
- Anna Zhukova
- Unité Bioinformatique Evolutive, Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI USR 3756 Institut Pasteur et CNRS, France
| | | | - Olivier Gascuel
- Unité Bioinformatique Evolutive, Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI USR 3756 Institut Pasteur et CNRS, France
| | - Deenan Pillay
- Division of Infection and Immunity, University College London, United Kingdom.,Africa Health Research Institute, KwaZulu-Natal, South Africa
| |
Collapse
|
41
|
Yerly S, Calmy A. Time to overcome pretreatment HIV drug resistance. THE LANCET. INFECTIOUS DISEASES 2017; 18:239-240. [PMID: 29198912 DOI: 10.1016/s1473-3099(17)30709-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 11/17/2022]
Affiliation(s)
- Sabine Yerly
- Laboratory of Virology, AIDS Unit, Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva 1211, Switzerland.
| | - Alexandra Calmy
- Laboratory of Virology, AIDS Unit, Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva 1211, Switzerland
| |
Collapse
|
42
|
Assoumou L, Charpentier C, Recordon-Pinson P, Grudé M, Pallier C, Morand-Joubert L, Fafi-Kremer S, Krivine A, Montes B, Ferré V, Bouvier-Alias M, Plantier JC, Izopet J, Trabaud MA, Yerly S, Dufayard J, Alloui C, Courdavault L, Le Guillou-Guillemette H, Maillard A, Amiel C, Vabret A, Roussel C, Vallet S, Guinard J, Mirand A, Beby-Defaux A, Barin F, Allardet-Servent A, Ait-Namane R, Wirden M, Delaugerre C, Calvez V, Chaix ML, Descamps D, Reigadas S. Prevalence of HIV-1 drug resistance in treated patients with viral load >50 copies/mL: a 2014 French nationwide study. J Antimicrob Chemother 2017; 72:1769-1773. [PMID: 28333232 DOI: 10.1093/jac/dkx042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/19/2017] [Indexed: 11/14/2022] Open
Abstract
Background Surveillance of HIV-1 resistance in treated patients with a detectable viral load (VL) is important to monitor, in order to assess the risk of spread of resistant viruses and to determine the proportion of patients who need new antiretroviral drugs with minimal cross-resistance. Methods The HIV-1 protease and reverse transcriptase (RT) and integrase genes were sequenced in plasma samples from 782 consecutive patients on failing antiretroviral regimens, seen in 37 specialized centres in 2014. The genotyping results were interpreted using the ANRS v24 algorithm. Prevalence rates were compared with those obtained during a similar survey conducted in 2009. Results The protease and RT sequences were obtained in 566 patients, and the integrase sequence in 382 patients. Sequencing was successful in 60%, 78%, 78% and 87% of patients with VLs of 51-200, 201-500, 501-1000 and >1000 copies/mL, respectively. Resistance to at least one antiretroviral drug was detected in 56.3% of samples. Respectively, 3.9%, 8.7%, 1.5% and 3.4% of patients harboured viruses that were resistant to any NRTI, NNRTI, PI and integrase inhibitor (INI). Resistance rates were lower in 2014 than in 2009. Resistance was detected in 48.5% of samples from patients with a VL between 51 and 200 copies/mL. Conclusion In France in 2014, 90.0% of patients in AIDS care centres were receiving antiretroviral drugs and 12.0% of them had VLs >50 copies/mL. Therefore, this study suggests that 6.7% of treated patients in France might transmit resistant strains. Resistance testing may be warranted in all treated patients with VL > 50 copies/mL.
Collapse
Affiliation(s)
- L Assoumou
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP UMRS 1136), F75013 Paris, France
| | - C Charpentier
- INSERM UMR1137, IAME Université Paris Diderot Sorbonne Paris Cité, AP-HP, Laboratoire de Virologie, Hôpital Bichat-Claude Bernard, Laboratoire Associé au Centre National de Référence du VIH-Résistance aux Antirétroviraux, Paris, France
| | - P Recordon-Pinson
- PTBM, Laboratoire de Virologie, Hôpital Pellegrin, CHU de Bordeaux; UMR 5234 MFP CNRS, Université de Bordeaux, 33076 Bordeaux cedex, France
| | - M Grudé
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP UMRS 1136), F75013 Paris, France
| | - C Pallier
- HU Paris sud, Hôpital Paul Brousse, Laboratoire de Virologie, Villejuif, France
| | - L Morand-Joubert
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP UMRS 1136), AP-HP, Laboratoire de Virologie, Hôpital Saint-Antoine, F75012 Paris, France
| | - S Fafi-Kremer
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - A Krivine
- AP-HP, Hôpital Cochin, Laboratoire de Virologie, Paris, France
| | - B Montes
- Laboratoire de Virologie, Hôpital Saint-Eloi, CHU Montpellier, Montpellier, France
| | - V Ferré
- EA 4271, Nantes Université UFR Pharmacie, Laboratoire de Virologie, CHU Nantes, Nantes, France
| | - M Bouvier-Alias
- INSERM U955, National Reference Center for Viral Hepatitis B, C et Delta, Department of Virology, Henri Mondor Hospital, University of Paris-Est, Créteil, France
| | - J-C Plantier
- Laboratoire de Virologie et COREVIH Haute-Normandie, CHU de Rouen, Rouen, France
| | - J Izopet
- Laboratoire de Virologie, Hôpital Purpan de Toulouse, Toulouse, France
| | - M-A Trabaud
- Laboratoire de Virologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - S Yerly
- Laboratoire de Virologie, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - J Dufayard
- Laboratoire de Virologie, Hôpital l'Archet de Nice, Nice, France
| | - C Alloui
- Laboratoire de Virologie, Hôpital Avicenne, APHP, HU Paris Seine Saint Denis, Bobigny, France
| | - L Courdavault
- Laboratoire de Virologie, Centre Hospitalier Victor Dupouy d'Argenteuil, Argenteuil, France
| | - H Le Guillou-Guillemette
- Laboratoire de Virologie, CHU Angers et HIFIH Laboratory, UPRES 3859, SFR 4208, LUNAM University, Angers, France
| | - A Maillard
- Laboratoire de Virologie, CHU de Rennes, Rennes, France
| | - C Amiel
- AP-HP, Hôpital Tenon, Laboratoire de Virologie, Paris, France
| | - A Vabret
- Laboratoire de Virologie, CHU Caen, Caen, France
| | - C Roussel
- Laboratoire de Virologie, CHU Amiens, Amiens, France
| | - S Vallet
- Laboratoire de Virologie, CHU Brest, Brest, France
| | - J Guinard
- Laboratoire de Virologie, CHR Orléans, Orléans, France
| | - A Mirand
- Laboratoire de Virologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - A Beby-Defaux
- Laboratoire de Virologie, CHU Poitiers, Poitiers, France
| | - F Barin
- Laboratoire de Virologie, CHU Bretonneau, & INSERM U966, Tours, France
| | | | - R Ait-Namane
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP UMRS 1136), F75013 Paris, France
| | - M Wirden
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, Institut Pierre Louis d'épidémiologie et de Santé Publique (IPLESP UMRS 1136), AP-HP, Laboratoire de Virologie, Hôpital Pitié-Salpêtrière, F75013 Paris, France
| | - C Delaugerre
- Laboratoire de Virologie, AP-HP, Hôpital Saint Louis, INSERM U941, Université Paris Diderot, Paris, France
| | - V Calvez
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, Institut Pierre Louis d'épidémiologie et de Santé Publique (IPLESP UMRS 1136), AP-HP, Laboratoire de Virologie, Hôpital Pitié-Salpêtrière, F75013 Paris, France
| | - M-L Chaix
- Laboratoire de Virologie, AP-HP, Hôpital Saint Louis, INSERM U941, Université Paris Diderot, Paris, France
| | - D Descamps
- INSERM UMR1137, IAME Université Paris Diderot Sorbonne Paris Cité, AP-HP, Laboratoire de Virologie, Hôpital Bichat-Claude Bernard, Laboratoire Associé au Centre National de Référence du VIH-Résistance aux Antirétroviraux, Paris, France
| | - S Reigadas
- PTBM, Laboratoire de Virologie, Hôpital Pellegrin, CHU de Bordeaux; UMR 5234 MFP CNRS, Université de Bordeaux, 33076 Bordeaux cedex, France.,CRB plurithématique, Bordeaux Biothèques Santé, Groupe hospitalier Pellegrin-CHU de Bordeaux, Bordeaux, France
| | | |
Collapse
|
43
|
Tsai HC, Chen IT, Wu KS, Tseng YT, Sy CL, Chen JK, Lee SSJ, Chen YS. High rate of HIV-1 drug resistance in treatment failure patients in Taiwan, 2009-2014. Infect Drug Resist 2017; 10:343-352. [PMID: 29081666 PMCID: PMC5652926 DOI: 10.2147/idr.s146584] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Drug resistance to nucleoside reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and protease inhibitors (PIs) has been associated with loss of viral suppression measured by a rise in HIV-1 RNA levels, a decline in CD4 cell counts, persistence on a failing treatment regimen, and lack of adherence to combination antiretroviral therapy. Objectives This study aimed to monitor the prevalence and risk factors associated with drug resistance in Taiwan after failure of first-line therapy. Materials and methods Data from the Veterans General Hospital Surveillance and Monitor Network for the period 2009–2014 were analyzed. Plasma samples from patients diagnosed with virologic failure and an HIV-1 RNA viral load >1000 copies/mL were analyzed by the ViroSeq™ HIV-1 genotyping system for drug susceptibility. Hazard ratios (HRs) for drug resistance were calculated using a Cox proportional hazard model. Results From 2009 to 2014, 359 patients were tested for resistance. The median CD4 count and viral load (log) were 214 cells/μL (interquartile range [IQR]: 71–367) and 4.5 (IQR: 3.9–5.0), respectively. Subtype B HIV-1 strains were found in 90% of individuals. The resistance rate to any of the three classes of antiretroviral drugs (NRTI, NNRTI, and PI) was 75.5%. The percentage of NRTI, NNRTI, and PI resistance was 58.6%, 61.4%, and 11.4%, respectively. The risk factors for any class of drug resistance included age ≤35 years (adjusted HR: 2.30, CI: 1.48–3.56; p<0.0001), initial NNRTI-based antiretroviral regimens (adjusted HR: 1.70, CI: 1.10–2.63; p=0.018), and current NNRTI-based antiretroviral regimens when treatment failure occurs (odds ratio: 4.04, CI: 2.47–6.59; p<0.001). There was no association between HIV-1 subtype, viral load, and resistance. Conclusion This study demonstrated a high level of resistance to NRTI and NNRTI in patients with virologic failure to first-line antiretroviral therapy despite routine viral load monitoring. Educating younger men who have sex with men to maintain good adherence is crucial, as PI use is associated with lower possibility of drug resistance.
Collapse
Affiliation(s)
- Hung-Chin Tsai
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei.,Department of Parasitology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Tzu Chen
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung
| | - Kuan-Sheng Wu
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei
| | - Yu-Ting Tseng
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung
| | - Cheng-Len Sy
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung
| | - Jui-Kuang Chen
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung
| | - Susan Shin-Jung Lee
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei
| | - Yao-Shen Chen
- Department of Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei
| |
Collapse
|
44
|
Temporal Patterns and Drug Resistance in CSF Viral Escape Among ART-Experienced HIV-1 Infected Adults. J Acquir Immune Defic Syndr 2017; 75:246-255. [PMID: 28328546 PMCID: PMC5452976 DOI: 10.1097/qai.0000000000001362] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Supplemental Digital Content is Available in the Text. Background: Cerebrospinal fluid (CSF) viral escape is an increasingly recognized clinical event among HIV-1-infected adults. We analyzed longitudinal data and drug-resistance mutations to characterize profiles of HIV-1-infected patients on antiretroviral therapy with discordant CSF and plasma HIV-1 RNA levels. Methods: Forty-one cases of CSF escape defined as detectable CSF HIV-1 RNA when plasma levels were undetectable, or HIV-1 RNA >0.5-log higher in CSF than plasma were identified from Boston Hospitals and National NeuroAIDS Tissue Consortium (NNTC) from 2005 to 2016. Results: Estimated prevalence of CSF escape in Boston and NNTC cohorts was 6.0% and 6.8%, respectively; median age was 50, duration of HIV-1 infection 17 years, CD4 count 329 cells/mm3 and CD4 nadir 21 cells/mm3. Neurological symptoms were present in 30 cases; 4 had repeat episodes of CSF escape. Cases were classified into subtypes based plasma HIV-1 RNA levels in the preceding 24 months: high-level viremia (1000 copies/mL), low-level viremia (LLV: 51–999 copies/mL), and plasma suppression with CSF blip or escape (CSF RNA <200 or ≥200 copies/mL). High-level viremia cases reported more substance abuse, whereas LLV or plasma suppression cases were more neurosymptomatic (81% vs. 53%); 75% of repeat CSF escape cases were classified LLV. M184V/I mutations were identified in 74% of CSF samples when plasma levels were ≤50 copies per milliliter. Conclusions: Characteristics frequently observed in CSF escape include HIV-1 infection >15 years, previous LLV, and M184V/I mutations in CSF. Classification based on preceding plasma HIV RNA levels provides a useful conceptual framework to identify causal factors and test therapeutics.
Collapse
|
45
|
Rocheleau G, Brumme CJ, Shoveller J, Lima VD, Harrigan PR. Longitudinal trends of HIV drug resistance in a large Canadian cohort, 1996-2016. Clin Microbiol Infect 2017; 24:185-191. [PMID: 28652115 DOI: 10.1016/j.cmi.2017.06.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 11/25/2022]
Abstract
OBJECTIVES We aim to identify long-term trends in HIV drug resistance before and after combined antiretroviral therapy (cART) initiation. METHODS IAS-USA (2015) mutations were identified in 23 271 HIV protease-reverse transcriptase sequences from 6543 treatment naïve adults in British Columbia. Participants who started cART between 1996 and 2014 were followed until April 2016. Equality of proportions test was used to compare the percentage of participants with acquired drug resistance (ADR) or transmitted drug resistance (TDR) in 1996, to those in 2014. Kaplan-Meier was used to estimate time to ADR in four drug resistance categories. Multivariable regression odds ratios (OR) of ADR for select clinical variables were determined by 5-year eras of cART initiation. RESULTS The proportion of individuals with ADR declined from 39% (51/132) to 3% (8/322) in 1996-2014 (p <0.0001), while the proportion with TDR increased from 12% (16/132) to 18% (59/322) (p 0.14). The estimated proportions of individuals with ADR rose to 29% (NNRTI), 28% (3TC/FTC), 14% (other nRTI), and 7% (PI) after >16 years of therapy. After 5 years on therapy, participants initiating cART in 1996-2000 had 5.5-times more 3TC/FTC ADR, 5.3-times more other nRTI ADR, 4.7-times more NNRTI ADR, and 24-times more PI ADR than those starting in 2011-2014. The individuals with highest odds of developing ADR in 1996-2010 were adherent to regimens at levels between 60% and 80%, which shifted to <40% adherent in 2011-2014. CONCLUSIONS HIV drug resistance transitioned from being primarily selected de-novo to being driven by TDR. Among those who started treatment in the past 5 years, ADR is rare and observed mostly in the lowest adherence strata.
Collapse
Affiliation(s)
- G Rocheleau
- Experimental Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada; British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - C J Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - J Shoveller
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada; School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - V D Lima
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada; School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - P R Harrigan
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada; Division of AIDS, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
46
|
Sepúlveda-Torres LDC, Rishishwar L, Rogers ML, Ríos-Olivares E, Boukli N, Jordan IK, Cubano LA. A decade of viral mutations and associated drug resistance in a population of HIV-1+ Puerto Ricans: 2002-2011. PLoS One 2017; 12:e0177452. [PMID: 28493944 PMCID: PMC5426751 DOI: 10.1371/journal.pone.0177452] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/27/2017] [Indexed: 12/14/2022] Open
Abstract
Puerto Rico has one of the highest rates of HIV/AIDS seen for any US state or territory, and antiretroviral therapy has been a mainstay of efforts to mitigate the HIV/AIDS public health burden on the island. We studied the evolutionary dynamics of HIV-1 mutation and antiretroviral drug resistance in Puerto Rico by monitoring the population frequency of resistance-associated mutations from 2002 to 2011. Whole blood samples from 4,475 patients were analyzed using the TRUGENE HIV-1 Genotyping Kit and OpenGene DNA Sequencing System in the Immunoretrovirus Research Laboratory at Universidad Central del Caribe. Results show that 64.0% of female and 62.9% of male patients had HIV-1 mutations that confer resistance to at least one antiretroviral medication. L63P and M184V were the dominant mutations observed for the protease (PRO) and reverse transcriptase (RT) encoding genes, respectively. Specific resistance mutations, along with their associated drug resistance profiles, can be seen to form temporal clusters that reveal a steadily changing landscape of resistance trends over time. Both women and men showed resistance mutations for an average of 4.8 drugs over the 10-year period, further underscoring the strong selective pressure exerted by antiretrovirals along with the rapid adaptive response of HIV. Nevertheless, both female and male patients showed a precipitous decrease for overall drug resistance, and for PRO mutations in particular, over the entire course of the study, with the most rapid decrease in frequency seen after 2006. The reduced HIV-1 mutation and drug resistance trends that we observed are consistent with previous reports from multi-year studies conducted around the world. Reduced resistance can be attributed to the use of more efficacious antiretroviral drug therapy, including the introduction of multi-drug combination therapies, which limited the ability of the virus to mount rapid adaptive responses to antiretroviral selection pressure.
Collapse
Affiliation(s)
- Lycely del C. Sepúlveda-Torres
- Department of Microbiology and Immunology, Universidad Central del Caribe, Bayamón, Puerto Rico, United States of America
| | - Lavanya Rishishwar
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- PanAmerican Bioinformatics Institute, Cali, Valle del Cauca, Colombia
- Applied Bioinformatics Laboratory, Atlanta, Georgia, United States of America
| | - Maria Luisa Rogers
- Natural Sciences Department, Sacred Heart University, San Juan, Puerto Rico, United States of America
| | - Eddy Ríos-Olivares
- Department of Microbiology and Immunology, Universidad Central del Caribe, Bayamón, Puerto Rico, United States of America
| | - Nawal Boukli
- Department of Microbiology and Immunology, Universidad Central del Caribe, Bayamón, Puerto Rico, United States of America
| | - I. King Jordan
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- PanAmerican Bioinformatics Institute, Cali, Valle del Cauca, Colombia
- Applied Bioinformatics Laboratory, Atlanta, Georgia, United States of America
| | - Luis A. Cubano
- Department of Microbiology and Immunology, Universidad Central del Caribe, Bayamón, Puerto Rico, United States of America
| |
Collapse
|
47
|
In-depth analysis of HIV-1 drug resistance mutations in HIV-infected individuals failing first-line regimens in West and Central Africa. AIDS 2016; 30:2577-2589. [PMID: 27603287 DOI: 10.1097/qad.0000000000001233] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE In resource-limited countries, antiretroviral therapy (ART) has been scaled up, but individual monitoring is still suboptimal. Here, we studied whether or not ART had an impact on the frequency and selection of drug resistance mutations (DRMs) under these settings. We also examined whether differences exist between HIV-1 genetic variants. DESIGN A total of 3736 sequences from individuals failing standard first-line ART (n = 1599, zidovudine/stavudine + lamivudine + neviparine/efavirenz) were analyzed and compared with sequences from reverse transcriptase inhibitor (RTI)-naive individuals (n = 2137) from 10 West and Central African countries. METHODS Fisher exact tests and corrections for multiple comparisons were used to assess the significance of associations. RESULTS All RTI-DRM from the 2015 International Antiviral Society list, except F227C, and nine mutations from other expert lists were observed to confer extensive resistance and cross-resistance. Five additional independently selected mutations (I94L, L109I, V111L, T139R and T165L) were statistically associated with treatment. The proportion of sequences with multiple mutations and the frequency of all thymidine analog mutations, M184V, certain NNRTIS, I94L and L109I showed substantial increase with time on ART. Only one nucleoside and two nonnucleoside RTI-DRMs differed by subtype/circulating recombinant form. CONCLUSION This study validates the global robustness of the actual DRM repertoire, in particular for circulating recombinant form 02 predominating in West and Central Africa, despite our finding of five additional selected mutations. However, long-term ART without virological monitoring clearly leads to the accumulation of mutations and the emergence of additional variations, which limit drug options for treatment and can be transmitted. Improved monitoring and optimization of ART are necessary for the long-term effectiveness of ART.
Collapse
|
48
|
Richman DD. Editorial Commentary: HIV Is Putting Up Less Resistance. Clin Infect Dis 2016; 62:1318-9. [PMID: 26962077 DOI: 10.1093/cid/ciw130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Douglas D Richman
- Departments of Pathology and Medicine and the Center for AIDS Research, University of California VA San Diego Healthcare System
| |
Collapse
|