101
|
Crowell TA, Danboise B, Parikh A, Esber A, Dear N, Coakley P, Kasembeli A, Maswai J, Khamadi S, Bahemana E, Iroezindu M, Kiweewa F, Owuoth J, Freeman J, Jagodzinski LL, Malia JA, Eller LA, Tovanabutra S, Peel SA, Ake JA, Polyak CS. Pretreatment and Acquired Antiretroviral Drug Resistance Among Persons Living With HIV in Four African Countries. Clin Infect Dis 2020; 73:e2311-e2322. [PMID: 32785695 PMCID: PMC8492117 DOI: 10.1093/cid/ciaa1161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Emerging HIV drug resistance (HIVDR) could jeopardize the success of standardized HIV management protocols in resource-limited settings. We characterized HIVDR among antiretroviral therapy (ART)-naive and experienced participants in the African Cohort Study (AFRICOS). METHODS From January 2013 to April 2019, adults with HIV-1 RNA >1000 copies/mL underwent ART history review and HIVDR testing upon enrollment at 12 clinics in Uganda, Kenya, Tanzania, and Nigeria. We calculated resistance scores for specific drugs and tallied major mutations to non-nucleoside reverse transcriptase inhibitors (NNRTIs), nucleoside reverse transcriptase inhibitors (NRTIs), and protease inhibitors (PIs) using Stanford HIVDB 8.8 and SmartGene IDNS software. For ART-naive participants, World Health Organization surveillance drug resistance mutations (SDRMs) were noted. RESULTS HIVDR testing was performed on 972 participants with median age 35.7 (interquartile range [IQR] 29.7-42.7) years and median CD4 295 (IQR 148-478) cells/mm3. Among 801 ART-naive participants, the prevalence of SDRMs was 11.0%, NNRTI mutations 8.2%, NRTI mutations 4.7%, and PI mutations 0.4%. Among 171 viremic ART-experienced participants, NNRTI mutation prevalence was 83.6%, NRTI 67.8%, and PI 1.8%. There were 90 ART-experienced participants with resistance to both efavirenz and lamivudine, 33 (36.7%) of whom were still prescribed these drugs. There were 10 with resistance to both tenofovir and lamivudine, 8 (80.0%) of whom were prescribed these drugs. CONCLUSIONS Participants on failing ART regimens had a high burden of HIVDR that potentially limited the efficacy of standardized first- and second-line regimens. Management strategies that emphasize adherence counseling while delaying ART switch may promote drug resistance and should be reconsidered.
Collapse
Affiliation(s)
- Trevor A Crowell
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Brook Danboise
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ajay Parikh
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Allahna Esber
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Nicole Dear
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Peter Coakley
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Alex Kasembeli
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,HJF Medical Research International, Kericho, Kenya
| | - Jonah Maswai
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,HJF Medical Research International, Kericho, Kenya
| | - Samoel Khamadi
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,HJF Medical Research International, Mbeya, Tanzania
| | - Emmanuel Bahemana
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,HJF Medical Research International, Mbeya, Tanzania
| | - Michael Iroezindu
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,HJF Medical Research International, Abuja, Nigeria
| | | | - John Owuoth
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,HJF Medical Research International, Kisumu, Kenya
| | - Joanna Freeman
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Linda L Jagodzinski
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jennifer A Malia
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Leigh Ann Eller
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Sodsai Tovanabutra
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Sheila A Peel
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Julie A Ake
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Christina S Polyak
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | | |
Collapse
|
102
|
Assoumou L, Bocket L, Pallier C, Grude M, Ait-Namane R, Izopet J, Raymond S, Charpentier C, Visseaux B, Wirden M, Trabaud MA, Le Guillou-Guillemette H, Allaoui C, Henquell C, Krivine A, Dos Santos G, Delamare C, Bouvier-Alias M, Montes B, Ferre V, De Monte A, Signori-Schmuck A, Maillard A, Morand-Joubert L, Tumiotto C, Fafi-Kremer S, Amiel C, Barin F, Marque-Juillet S, Courdavault L, Vallet S, Beby-Defaux A, de Rougemont A, Fenaux H, Avettand-Fenoel V, Allardet-Servent A, Plantier JC, Peytavin G, Calvez V, Chaix ML, Descamps D. Stable prevalence of transmitted drug resistance mutations and increased circulation of non-B subtypes in antiretroviral-naive chronically HIV-infected patients in 2015/2016 in France. J Antimicrob Chemother 2020; 74:1417-1424. [PMID: 30753724 DOI: 10.1093/jac/dkz011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/27/2018] [Accepted: 12/31/2018] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES We estimated the prevalence of transmitted-drug-resistance-associated mutations (TDRAMs) in antiretroviral-naive chronically HIV-1-infected patients. PATIENTS AND METHODS TDRAMs were sought in samples from 660 diagnosed HIV-1-infected individuals in 2015/2016 in 33 HIV clinical centres. Weighted analyses, considering the number of patients followed in each centre, were used to derive representative estimates of the percentage of individuals with TDRAMs. Results were compared with those of the 2010/2011 survey (n = 661) using the same methodology. RESULTS At inclusion, median CD4 cell counts and plasma HIV-1 RNA were 394 and 350/mm3 (P = 0.056) and 4.6 and 4.6 log10 copies/mL (P = 0.360) in the 2010/2011 survey and the 2015/2016 survey, respectively. The frequency of non-B subtypes increased from 42.9% in 2010/2011 to 54.8% in 2015/2016 (P < 0.001), including 23.4% and 30.6% of CRF02_AG (P = 0.004). The prevalence of virus with protease or reverse-transcriptase TDRAMs was 9.0% (95% CI = 6.8-11.2) in 2010/2011 and 10.8% (95% CI = 8.4-13.2) in 2015/2016 (P = 0.269). No significant increase was observed in integrase inhibitor TDRAMs (6.7% versus 9.2%, P = 0.146). Multivariable analysis showed that men infected with the B subtype were the group with the highest risk of being infected with a resistant virus compared with others (adjusted OR = 2.2, 95% CI = 1.3-3.9). CONCLUSIONS In France in 2015/2016, the overall prevalence of TDRAMs was 10.8% and stable compared with 9.0% in the 2010/2011 survey. Non-B subtypes dramatically increased after 2010. Men infected with B subtype were the group with the highest risk of being infected with a resistant virus, highlighting the need to re-emphasize safe sex messages.
Collapse
Affiliation(s)
- Lambert Assoumou
- INSERM, UMR 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France
| | | | | | - Maxime Grude
- INSERM, UMR 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France
| | - Rachid Ait-Namane
- INSERM, UMR 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France
| | | | | | - Charlotte Charpentier
- Hopital Bichat Claude Bernard, Virology, Paris, France.,Univ Paris-Diderot, INSERM UMR 1137, CNR VIH, Paris, France
| | - Benoit Visseaux
- Hopital Bichat Claude Bernard, Virology, Paris, France.,Univ Paris-Diderot, INSERM UMR 1137, CNR VIH, Paris, France
| | - Marc Wirden
- INSERM, UMR 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France.,CHU Pitié-Salpêtrière, Virology, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Laurence Morand-Joubert
- INSERM, UMR 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France.,Hopital Saint-Antoine, Virology, Paris, France.,Sorbonne Université, Paris, France
| | | | | | | | | | | | | | | | | | | | | | - Véronique Avettand-Fenoel
- CHU Necker-Enfants Malades, Virology, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | | | - Gilles Peytavin
- Univ Paris-Diderot, INSERM UMR 1137, CNR VIH, Paris, France.,Hopital Bichat-Claude Bernard, Laboratoire de Pharmaco-Toxicologie, Paris, France
| | - Vincent Calvez
- INSERM, UMR 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France.,CHU Pitié-Salpêtrière, Virology, Paris, France.,Sorbonne Université, Paris, France
| | | | - Diane Descamps
- Hopital Bichat Claude Bernard, Virology, Paris, France.,Univ Paris-Diderot, INSERM UMR 1137, CNR VIH, Paris, France
| | | |
Collapse
|
103
|
Gachogo RW, Mwai DN, Onyambu FG. Cost analysis of implementing HIV drug resistance testing in Kenya: a case study of a service delivery site at a tertiary level hospital in Kenya. F1000Res 2020; 9:793. [PMID: 32983418 PMCID: PMC7495211 DOI: 10.12688/f1000research.23379.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/15/2020] [Indexed: 01/13/2023] Open
Abstract
Background: HIV drug resistance (HIVDR) threatens progress achieved in response to the HIV epidemic. Understanding the costs of implementing HIVDR testing programs for patient management and surveillance in resource-limited settings is critical in optimizing resource allocation. Here, we estimate the unit cost of HIVDR testing and identify major cost drivers while documenting challenges and lessons learnt in implementation of HIVDR testing at a tertiary level hospital in Kenya. Methods: We employed a mixed costing approach to estimate the costs associated with performing a HIVDR test from the provider's perspective. Data collection involved a time and motion study of laboratory procedures and interviewing laboratory personnel and the management personnel. Cost analysis was based on estimated 1000 HIVDR tests per year. Data entry and analysis were done using Microsoft Excel and costs converted to US dollars (2019). Results: The estimated unit cost for a HIVDR test was $271.78 per test. The main cost drivers included capital ($102.42, 37.68%) and reagents (101.50, 37.35%). Other costs included: personnel ($46.81, 17.22%), utilities ($14.69, 5.41%), equipment maintenance costs ($2.37, 0.87%) and quality assurance program ($4, 1.47%). Costs in relation to specific laboratory processes were as follows: sample collection ($2.41, 0.89%), RNA extraction ($22.79, 8.38%), amplification ($56.14, 20.66%), gel electrophoresis ($10.34, 3.80%), sequencing ($160.94, 59.22%), and sequence analysis ($19.16, 7.05%). A user-initiated modification of halving reagent volumes for some laboratory processes (amplification and sequencing) reduced the unit cost for a HIVDR test to $233.81 (13.97%) reduction. Conclusions: Capital expenditure and reagents remain the most expensive components of HIVDR testing. This cost is bound to change as the sequencing platform is utilized towards maximum capacity or leveraged for use with other tests. Cost saving in offering HIVDR testing services is also possible through reagent volume reduction without compromising on the quality of test results.
Collapse
Affiliation(s)
- Rachael W. Gachogo
- Molecular and Infectious Diseases Research Laboratory, University of Nairobi, Nairobi, Kenya
- School of Economics, University of Nairobi, Nairobi, Kenya
| | - Daniel N. Mwai
- School of Economics, University of Nairobi, Nairobi, Kenya
| | - Frank G. Onyambu
- Molecular and Infectious Diseases Research Laboratory, University of Nairobi, Nairobi, Kenya
- School of Health Sciences, Meru University of Science and Technology, Meru, Kenya
| |
Collapse
|
104
|
Lathouwers E, Seyedkazemi S, Luo D, Brown K, De Meyer S, Wong EY. Pooled resistance analyses of darunavir once-daily regimens and formulations across 10 clinical studies of treatment-naïve and treatment-experienced patients with human immunodeficiency virus-1 infection. HIV Res Clin Pract 2020; 21:83-89. [PMID: 32715952 DOI: 10.1080/25787489.2020.1794439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND The efficacy and high barrier to resistance of darunavir have been demonstrated across diverse populations with HIV-1 infection. OBJECTIVE To evaluate post-baseline resistance among patients in studies of once-daily (QD) darunavir-based regimens and formulations. METHODS The analysis included treatment-naïve and virologically failing or suppressed patients from 10 phase 2/3 studies (48-192 weeks in duration) of boosted darunavir 800 mg QD-based regimens. Three were phase 3 studies of the QD darunavir/cobicistat/emtricitabine/tenofovir alafenamide (D/C/F/TAF) 800/150/200/10 mg single-tablet regimen. Post-baseline resistance was evaluated upon protocol-defined virologic failure (PDVF). Resistance-associated mutations (RAMs) were identified using International Antiviral Society-USA mutation lists. Phenotypic analyses varied across studies. RESULTS Overall, 250 of 3635 patients in the analysis met PDVF criteria; 205 had post-baseline genotypes/phenotypes. In total, four (0.1%) patients developed (or had identified) ≥1 darunavir and/or primary protease inhibitor (PI) RAM; only one (<0.1%) patient (with prior lopinavir virologic failure) lost darunavir phenotypic susceptibility. Among 3317 patients using nucleos(t)ide reverse transcriptase inhibitors (N[t]RTIs; mostly emtricitabine and tenofovir), 13 (0.4%) had ≥1 N(t)RTI RAM (10 with M184I/V). Among patients receiving D/C/F/TAF (n = 1949), none had post-baseline darunavir, primary PI, or tenofovir RAMs; only two (0.1%) patients developed an emtricitabine RAM, M184V/I. CONCLUSIONS Across a large, diverse population using darunavir 800 mg QD-based regimens and formulations, resistance development remains rare. After clinical trials that span >10 years, loss of phenotypic susceptibility to darunavir was only observed once in a PI-experienced patient and has never been observed in treatment-naïve patients, treatment-experienced PI-naïve patients, or treatment-experienced virologically suppressed patients.
Collapse
Affiliation(s)
| | | | - Donghan Luo
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | | | | | - Eric Y Wong
- Janssen Scientific Affairs, LLC, Titusville, NJ, USA
| |
Collapse
|
105
|
Chahine EB, Durham SH. Ibalizumab: The First Monoclonal Antibody for the Treatment of HIV-1 Infection. Ann Pharmacother 2020; 55:230-239. [PMID: 32659101 DOI: 10.1177/1060028020942218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To review the efficacy and safety of ibalizumab (IBA) in the treatment of HIV-1 infection. DATA SOURCES A literature search was performed using PubMed and Google Scholar (2010 to mid-June 2020) with the search terms TMB-355, TNX-355, and ibalizumab. Other resources included abstracts presented at recent conferences and the manufacturer's website and prescribing information. STUDY SELECTION AND DATA EXTRACTION All relevant English-language articles of studies assessing the efficacy and safety of IBA were included. DATA SYNTHESIS IBA is a monoclonal antibody that blocks HIV-1 from infecting CD4+ T cells. IBA is approved by the Food and Drug Administration, in combination with other antiretrovirals (ARVs), for the treatment of HIV-1 infection in heavily treatment-experienced adults with multidrug-resistant (MDR) HIV-1 infection failing their current ARVs. IBA demonstrated significant and sustained antiviral activity in patients with MDR HIV-1 infection who had advanced disease and limited treatment options. It carries a warning regarding the development of immune reconstitution inflammatory syndrome. Common adverse reactions include diarrhea, dizziness, nausea, and rash. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE IBA represents an attractive option for treatment-experienced adults with advanced HIV-1 infection who are no longer able to achieve viral suppression on oral ARV therapy alone and who are able to adhere to an infusion therapy every 2 weeks. As with other biologics, there is a potential for the development of antibodies to IBA that can compromise its efficacy and safety. CONCLUSION IBA provides a needed treatment option to achieve and maintain viral suppression in heavily treatment-experienced adults with MDR HIV-1 infection.
Collapse
Affiliation(s)
- Elias B Chahine
- Palm Beach Atlantic University Lloyld L. Gregory School of Pharmacy, West Palm Beach, FL, USA
| | | |
Collapse
|
106
|
Vannappagari V, Ragone L, Henegar C, van Wyk J, Brown D, Demarest J, Quercia R, St Clair M, Underwood M, Gatell JM, de Ruiter A, Aboud M. Prevalence of pretreatment and acquired HIV-1 mutations associated with resistance to lamivudine or rilpivirine: a systematic review. Antivir Ther 2020; 24:393-404. [PMID: 31503008 DOI: 10.3851/imp3331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Pretreatment and acquired drug resistance mutations (DRMs) can limit antiretroviral therapy effectiveness. METHODS We review prevalence of DRMs with resistance to nucleoside reverse transcriptase inhibitors (NRTIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs), focusing on lamivudine and rilpivirine, from 127 articles with >100,000 individuals with HIV-1 infection. RESULTS Estimated global prevalence of pretreatment resistance to any NRTI was 4% and to any NNRTI was 6%. Most prevalent DRMs resistant to lamivudine or rilpivirine were at positions E138 (4%), V179 (1%) and M184 (1%). Estimated acquired DRM prevalence was 58% for any NRTIs and 67% for any NNRTIs, most frequently at positions M184 (58%) and Y181 (21%). CONCLUSIONS This review suggests low risk of lamivudine- or rilpivirine-resistant mutations in treatment-naive, HIV-1-infected individuals.
Collapse
Affiliation(s)
| | - Leigh Ragone
- ViiV Healthcare, Research Triangle Park, NC, USA
| | | | | | | | | | | | | | | | - Jose M Gatell
- Hospital Clinic/IDIBAPS, University of Barcelona, Barcelona, Spain.,ViiV Healthcare, Barcelona, Spain
| | | | | |
Collapse
|
107
|
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
|
108
|
Patil A, Elwitigala JP, Rajapaksa L, Gangakhedkar R, Chaturbhuj D, Pendse R, Rajapaksha DI, Rewari BB, Malliawadu N, Jayamanna K, Dombawela D, Kurle S. HIV-1 pol gene diversity and molecular dating of subtype C from Sri Lanka. PLoS One 2020; 15:e0234133. [PMID: 32525892 PMCID: PMC7289380 DOI: 10.1371/journal.pone.0234133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/19/2020] [Indexed: 11/19/2022] Open
Abstract
Background The first case of HIV infection in Sri Lanka was reported in 1987 and at the end of 2018 there were 3500 people living with HIV. There have been commendable efforts made towards the detection, treatment, and prevention of HIV in the country. Even though the genetic diversity of HIV has been shown to affect the parameters ranging from detection to vaccine development, there is no data available with respect to the molecular epidemiology of HIV-1 in Sri Lanka. Methods In this report we have performed the ancillary analysis of pol gene region sequences (n = 85) obtained primarily for the purpose of HIV-1 drug resistance genotyping. Briefly, dried blood spot specimens (DBS) collected from HIV-1 infected individuals between December 2015 and August 2018 were subjected to pol gene amplification and sequencing. These pol gene sequences were used to interpret the drug resistance mutation profiles. Further, sequences were subjected to HIV-1 subtyping using REGA 3.0, COMET, jPHMM and, RIP online subtyping tools. Moreover, Bayesian phylogenetic analysis was employed to estimate the evolutionary history of HIV-1 subtype C in Sri Lanka. Results Our analysis revealed that the majority (51.8%) of pol gene sequences were subtype C. Other than subtype C, there were sequences categorized as subtypes A1, B, D and G. In addition to pure subtypes there were sequences which were observed to be circulating recombinant forms (CRFs) and a few of the recombinants were identified as potential unique recombinants (URFs). We also observed the presence of drug resistance mutations in 56 (65.9%) out of 85 sequences. Estimates of the Bayesian evolutionary analysis suggested that the HIV-1 subtype C was introduced to Sri Lanka during the early 1970s (1972.8). Conclusion The findings presented here indicate the presence of multiple HIV-1 subtypes and the prevalence of drug resistance mutations in Sri Lanka. The majority of the sequences were subtype C, having their most recent common ancestor traced back to the early 1970s. Continuous molecular surveillance of HIV-1 molecular epidemiology will be crucial to keep track of drug resistance, genetic diversity, and evolutionary history of HIV-1 in Sri Lanka.
Collapse
Affiliation(s)
- Ajit Patil
- HIV Drug Resistance Laboratory, National AIDS Research Institute, Pune, India
| | | | | | | | - Devidas Chaturbhuj
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | | | | | - B. B. Rewari
- World Health Organization South-East Asia Region, New Delhi, India
| | | | | | | | - Swarali Kurle
- HIV Drug Resistance Laboratory, National AIDS Research Institute, Pune, India
- * E-mail:
| |
Collapse
|
109
|
Becker MG, Liang D, Cooper B, Le Y, Taylor T, Lee ER, Wu S, Sandstrom P, Ji H. Development and Application of Performance Assessment Criteria for Next-Generation Sequencing-Based HIV Drug Resistance Assays. Viruses 2020; 12:E627. [PMID: 32532083 PMCID: PMC7354553 DOI: 10.3390/v12060627] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/29/2020] [Accepted: 06/07/2020] [Indexed: 12/19/2022] Open
Abstract
Next-generation sequencing (NGS)-based HIV drug resistance (HIVDR) assays outperform conventional Sanger sequencing in scalability, sensitivity, and quantitative detection of minority resistance variants. Thus far, HIVDR assays have been applied primarily in research but rarely in clinical settings. One main obstacle is the lack of standardized validation and performance evaluation systems that allow regulatory agencies to benchmark and accredit new assays for clinical use. By revisiting the existing principles for molecular assay validation, here we propose a new validation and performance evaluation system that helps to both qualitatively and quantitatively assess the performance of an NGS-based HIVDR assay. To accomplish this, we constructed a 70-specimen proficiency test panel that includes plasmid mixtures at known ratios, viral RNA from infectious clones, and anonymized clinical specimens. We developed assessment criteria and benchmarks for NGS-based HIVDR assays and used these to assess data from five separate MiSeq runs performed in two experienced HIVDR laboratories. This proposed platform may help to pave the way for the standardization of NGS HIVDR assay validation and performance evaluation strategies for accreditation and quality assurance purposes in both research and clinical settings.
Collapse
Affiliation(s)
- Michael G. Becker
- National HIV and Retrovirology Laboratories, National Microbiology Laboratory at JC Wilt Infectious Diseases Research Center, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.G.B.); (T.T.); (E.R.L.); (P.S.)
| | - Dun Liang
- ViroDx Clinical Diagnostics Laboratory, St. Louis, MO 63017, USA; (D.L.); (B.C.); (Y.L.)
| | - Breanna Cooper
- ViroDx Clinical Diagnostics Laboratory, St. Louis, MO 63017, USA; (D.L.); (B.C.); (Y.L.)
| | - Yan Le
- ViroDx Clinical Diagnostics Laboratory, St. Louis, MO 63017, USA; (D.L.); (B.C.); (Y.L.)
| | - Tracy Taylor
- National HIV and Retrovirology Laboratories, National Microbiology Laboratory at JC Wilt Infectious Diseases Research Center, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.G.B.); (T.T.); (E.R.L.); (P.S.)
| | - Emma R. Lee
- National HIV and Retrovirology Laboratories, National Microbiology Laboratory at JC Wilt Infectious Diseases Research Center, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.G.B.); (T.T.); (E.R.L.); (P.S.)
| | - Sutan Wu
- SutanStats, St. Louis, MO 63017, USA;
| | - Paul Sandstrom
- National HIV and Retrovirology Laboratories, National Microbiology Laboratory at JC Wilt Infectious Diseases Research Center, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.G.B.); (T.T.); (E.R.L.); (P.S.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Hezhao Ji
- National HIV and Retrovirology Laboratories, National Microbiology Laboratory at JC Wilt Infectious Diseases Research Center, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.G.B.); (T.T.); (E.R.L.); (P.S.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
110
|
Courlet P, Alves Saldanha S, Cavassini M, Marzolini C, Choong E, Csajka C, Günthard HF, André P, Buclin T, Desfontaine V, Decosterd LA. Development and validation of a multiplex UHPLC-MS/MS assay with stable isotopic internal standards for the monitoring of the plasma concentrations of the antiretroviral drugs bictegravir, cabotegravir, doravirine, and rilpivirine in people living with HIV. JOURNAL OF MASS SPECTROMETRY : JMS 2020; 55:e4506. [PMID: 32160389 PMCID: PMC7317362 DOI: 10.1002/jms.4506] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/04/2020] [Accepted: 02/15/2020] [Indexed: 06/04/2023]
Abstract
The widespread use of highly active antiretroviral treatments has dramatically changed the prognosis of people living with HIV (PLWH). However, such treatments have to be taken lifelong raising issues regarding the maintenance of both therapeutic effectiveness and long-term tolerability. Recently approved or investigational antiretroviral drugs present considerable advantages, allowing once daily oral dosage along with activity against resistant variants (eg, bictegravir and doravirine) and also parenteral intramuscular administration that facilitates treatment adherence (eg, long-acting injectable formulations such as cabotegravir and rilpivirine). Still, there remains a risk of insufficient or exaggerated circulating exposure due to absorption issues, abnormal elimination, drug-drug interactions, and others. In this context, a multiplex ultra-high performance liquid chromatography coupled to tandem mass spectrometry (UHPLC-MS/MS) bioassay has been developed for the monitoring of plasma levels of bictegravir, cabotegravir, doravirine, and rilpivirine in PLWH. A simple and convenient protein precipitation was performed followed by direct injection of the supernatant into the UHPLC-MS/MS system. The four analytes were eluted in less than 3 minutes using a reversed-phase chromatography method coupled with triple quadrupole mass spectrometry detection. This bioassay was fully validated following international guidelines and achieved good performances in terms of trueness (94.7%-107.5%), repeatability (2.6%-11%), and intermediate precision (3.0%-11.2%) over the clinically relevant concentration ranges (from 30 to 9000 ng/mL for bictegravir, cabotegravir, and doravirine and from 10 to 1800 ng/mL for rilpivirine). This sensitive, accurate, and rapid UHPLC-MS/MS assay is currently applied in our laboratory for routine therapeutic drug monitoring of the oral drugs bictegravir and doravirine and is also intended to be applied for the monitoring of cabotegravir/rilpivirine levels in plasma from PLWH receiving once monthly or every 2-month intramuscular injection of these long-acting antiretroviral drugs.
Collapse
Affiliation(s)
- Perrine Courlet
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Susana Alves Saldanha
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Matthias Cavassini
- Service of Infectious DiseasesLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Catia Marzolini
- Division of Infectious Diseases and Hospital EpidemiologyUniversity Hospital of BaselBaselSwitzerland
- University of BaselBaselSwitzerland
| | - Eva Choong
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Chantal Csajka
- Centre for Research and Innovation in Clinical Pharmaceutical SciencesUniversity Hospital and University of LausanneLausanneSwitzerland
- Institute of Pharmaceutical Sciences of Western SwitzerlandUniversity of Geneva, University of LausanneGenevaSwitzerland
- School of Pharmaceutical SciencesUniversity of GenevaGenevaSwitzerland
| | - Huldrych F. Günthard
- Division of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichSwitzerland
- Institute of Medical Virology, Swiss National Reference Centre for RetrovirusesUniversity of ZurichZurichSwitzerland
| | - Pascal André
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Thierry Buclin
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Vincent Desfontaine
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Laurent Arthur Decosterd
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| |
Collapse
|
111
|
Ji H, Sandstrom P, Paredes R, Harrigan PR, Brumme CJ, Avila Rios S, Noguera-Julian M, Parkin N, Kantor R. Are We Ready for NGS HIV Drug Resistance Testing? The Second "Winnipeg Consensus" Symposium. Viruses 2020; 12:E586. [PMID: 32471096 PMCID: PMC7354487 DOI: 10.3390/v12060586] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/13/2020] [Accepted: 05/25/2020] [Indexed: 12/31/2022] Open
Abstract
HIV drug resistance is a major global challenge to successful and sustainable antiretroviral therapy. Next-generation sequencing (NGS)-based HIV drug resistance (HIVDR) assays enable more sensitive and quantitative detection of drug-resistance-associated mutations (DRMs) and outperform Sanger sequencing approaches in detecting lower abundance resistance mutations. While NGS is likely to become the new standard for routine HIVDR testing, many technical and knowledge gaps remain to be resolved before its generalized adoption in regular clinical care, public health, and research. Recognizing this, we conceived and launched an international symposium series on NGS HIVDR, to bring together leading experts in the field to address these issues through in-depth discussions and brainstorming. Following the first symposium in 2018 (Winnipeg, MB Canada, 21-22 February, 2018), a second "Winnipeg Consensus" symposium was held in September 2019 in Winnipeg, Canada, and was focused on external quality assurance strategies for NGS HIVDR assays. In this paper, we summarize this second symposium's goals and highlights.
Collapse
Affiliation(s)
- Hezhao Ji
- National HIV and Retrovirology Laboratories at JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada;
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Paul Sandstrom
- National HIV and Retrovirology Laboratories at JC Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada;
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Roger Paredes
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, s/n, 08916 Badalona, Catalonia, Spain; (R.P.); (M.N.-J.)
- Infectious Diseases Department, Hospital Germans Trias i Pujol, 08916 Badalona, Catalonia, Spain
| | - P. Richard Harrigan
- Division of AIDS, Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
| | - Chanson J. Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, BC V6Z 1Y6, Canada;
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Santiago Avila Rios
- Centre for Research in Infectious Diseases, National Institute of Respiratory Diseases, Mexico City 14080, Mexico;
| | - Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, s/n, 08916 Badalona, Catalonia, Spain; (R.P.); (M.N.-J.)
- Chair in AIDS and Related Illnesses, Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic–Central University of Catalonia (UVic–UCC), Can Baumann, Ctra. de Roda, 70, 08500 Vic, Spain
| | - Neil Parkin
- Data First Consulting Inc., Sebastopol, CA 95472, USA;
| | - Rami Kantor
- Division of Infectious Diseases, Brown University Alpert Medical School, Providence, RI 02906, USA;
| |
Collapse
|
112
|
Pimentel V, Pingarilho M, Alves D, Diogo I, Fernandes S, Miranda M, Pineda-Peña AC, Libin P, Martins MRO, Vandamme AM, Camacho R, Gomes P, Abecasis A. Molecular Epidemiology of HIV-1 Infected Migrants Followed up in Portugal: Trends between 2001-2017. Viruses 2020; 12:v12030268. [PMID: 32121161 PMCID: PMC7150888 DOI: 10.3390/v12030268] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 11/16/2022] Open
Abstract
Migration is associated with HIV-1 vulnerability. Objectives: To identify long-term trends in HIV-1 molecular epidemiology and antiretroviral drug resistance (ARV) among migrants followed up in Portugal Methods: 5177 patients were included between 2001 and 2017. Rega, Scuel, Comet, and jPHMM algorithms were used for subtyping. Transmitted drug resistance (TDR) and Acquired drug resistance (ADR) were defined as the presence of surveillance drug resistance mutations (SDRMs) and as mutations of the IAS-USA 2015 algorithm, respectively. Statistical analyses were performed. Results: HIV-1 subtypes infecting migrants were consistent with the ones prevailing in their countries of origin. Over time, overall TDR significantly increased and specifically for Non-nucleoside reverse transcriptase inhibitor (NNRTIs) and Nucleoside reverse transcriptase inhibitor (NRTIs). TDR was higher in patients from Mozambique. Country of origin Mozambique and subtype B were independently associated with TDR. Overall, ADR significantly decreased over time and specifically for NRTIs and Protease Inhibitors (PIs). Age, subtype B, and viral load were independently associated with ADR. Conclusions: HIV-1 molecular epidemiology in migrants suggests high levels of connectivity with their country of origin. The increasing levels of TDR in migrants could indicate an increase also in their countries of origin, where more efficient surveillance should occur.
Collapse
Affiliation(s)
- Victor Pimentel
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
| | - Marta Pingarilho
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
| | - Daniela Alves
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
| | - Isabel Diogo
- Laboratório de Biologia Molecular (LMCBM, SPC, CHLO-HEM), 1349-019 Lisboa, Portugal; (I.D.); (S.F.); (P.G.)
| | - Sandra Fernandes
- Laboratório de Biologia Molecular (LMCBM, SPC, CHLO-HEM), 1349-019 Lisboa, 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), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
| | - Andrea-Clemencia Pineda-Peña
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia, Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111321, Colombia
| | - Pieter Libin
- KU Leuven, Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (P.L.); (R.C.)
- Artificial Intelligence lab, Department of computer science, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - M. Rosário O. Martins
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
| | - Anne-Mieke Vandamme
- Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical/Universidade Nova de Lisboa (IHMT/UNL), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
- KU Leuven, Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (P.L.); (R.C.)
| | - Ricardo Camacho
- KU Leuven, Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium; (P.L.); (R.C.)
| | - Perpétua Gomes
- Laboratório de Biologia Molecular (LMCBM, SPC, CHLO-HEM), 1349-019 Lisboa, 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), 1349-008 Lisboa, Portugal; (V.P.); (M.P.); (D.A.); (M.M.); (A.-C.P.-P.); (M.R.O.M.); (A.-M.V.)
- Correspondence:
| |
Collapse
|
113
|
Zhang T, Ding H, An M, Wang X, Tian W, Zhao B, Han X. Factors associated with high-risk low-level viremia leading to virologic failure: 16-year retrospective study of a Chinese antiretroviral therapy cohort. BMC Infect Dis 2020; 20:147. [PMID: 32066392 PMCID: PMC7026956 DOI: 10.1186/s12879-020-4837-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/30/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Low level viremia (LLV) often occurs during antiretroviral therapy (ART) against HIV-1. However, whether LLV increases the risk of virologic failure (VF) is controversial because of the non-uniform definitions of LLV and VF. METHODS A long-term first line regimen ART cohort from 2002 to 2018 from Shenyang, northeast China, was retrospectively studied. All participants were followed up every 3 to 6 months to evaluate the treatment effect. The high-risk LLV subgroups leading to VF (with strict standards) were explored with Cox proportional hazards model and linear mixed-effect model. The association factors of high-risk LLV were further explored using multivariate logistic regression analyses. RESULTS A total of 2155 HIV-1 infected participants were included; of these, 38.7% showed LLV. Both high level LLV (HLLV) and any other level LLV coupled with high level blip (HLB) showed higher risk of VF (hazards ratios, HRHLLV = 5.93, and HRHLB = 2.84, p < 0.05 respectively). Moreover, HR increased with prolonged duration of LLV. Independent factors associated with high-risk LLV included the zenith baseline viral load (VL) above 6 log copies/ml (aOR = 3.49, p = 0.002), nadir baseline CD4 + T cell counts below 200 cells/mm3 (aOR = 1.78, p = 0.011), Manchu (aOR = 2.03, p = 0.003), ART over 60 months (aOR = 1.81, p = 0.004), AZT + 3TC + NVP (aOR = 2.26, p < 0.001) or DDI-based regimen (aOR = 9.96, p = 0.002), and subtype B' infection (aOR = 8.22, p = 0.001). CONCLUSIONS In case of VF with strict standards, high-risk LLV leading to VF includes VL above 400 copies/ml, occurring at least once. Serious laboratory indicators or advanced stage of infection, long term ART and subtype B' infection might also predict the occurrence of high-risk LLV.
Collapse
Affiliation(s)
- Tong Zhang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Haibo Ding
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Minghui An
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Xiaonan Wang
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Wen Tian
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Bin Zhao
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China
| | - Xiaoxu Han
- NHC Key Laboratory of AIDS Immunology (China Medical University), Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, No 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China. .,National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China. .,Key Laboratory of AIDS Immunology of Liaoning Province, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China. .,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003, China.
| |
Collapse
|
114
|
HIV-1 Coreceptor Usage and Variable Loop Contact Impact V3 Loop Broadly Neutralizing Antibody Susceptibility. J Virol 2020; 94:JVI.01604-19. [PMID: 31694950 DOI: 10.1128/jvi.01604-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/22/2019] [Indexed: 12/24/2022] Open
Abstract
In clinical trials, HIV-1 broadly neutralizing antibodies (bnAbs) effectively lower plasma viremia and delay virus reemergence. The presence of less neutralization-susceptible strains prior to treatment decreases the efficacy of these antibody-based treatments, but neutralization sensitivity often cannot be predicted by sequence analysis alone. We found that phenotypically confirmed CXCR4-utilizing strains are less neutralization sensitive, especially to variable loop 3 (V3 loop)-directed bnAbs, than exclusively CCR5-utilizing strains in some, but not all, cases. Homology modeling suggested that the primary V3 loop bnAb epitope is equally accessible among CCR5- and CXCR4-using strains, although variants that exclusively use CXCR4 have V3 loop protrusions that interfere with CCR5 receptor interactions. Homology modeling also showed that among some, but not all, envelopes with decreased neutralization sensitivity, V1 loop orientation interfered with V3 loop-directed bnAb binding. Thus, there are likely different structural reasons for the coreceptor usage restriction and the different bnAb susceptibilities. Importantly, we show that individuals harboring envelopes with higher likelihood of using CXCR4 or greater predicted V1 loop interference have faster virus rebound and a lower maximum decrease in plasma viremia, respectively, after treatment with a V3 loop bnAb. Knowledge of receptor usage and homology models may be useful in developing future algorithms that predict treatment efficacy with V3 loop bnAbs.IMPORTANCE The efficacy of HIV-1 broadly neutralizing antibody (bnAb) therapies may be compromised by the preexistence of less susceptible variants. Sequence-based methods are needed to predict pretreatment variants' neutralization sensitivities. HIV-1 strains that exclusively use the CXCR4 receptor rather than the CCR5 receptor are less neutralization susceptible, especially to variable loop 3 (V3 loop) bnAbs in some, but not all, instances. While the inability to utilize the CCR5 receptor maps to a predicted protrusion in the envelope V3 loop, this viral determinant does not directly influence V3 loop bnAb sensitivity. Homology modeling predicts that contact between the envelope V1 loop and the antibody impacts V3 loop bnAb susceptibility in some cases. Among pretreatment envelopes, increased probability of using CXCR4 and greater predicted V1 interference are associated with faster virus rebound and a smaller decrease in the plasma virus level, respectively, after V3 loop bnAb treatment. Receptor usage information and homology models may be useful for predicting V3 loop bnAb therapy efficacy.
Collapse
|
115
|
Quasispecies dynamics in disease prevention and control. VIRUS AS POPULATIONS 2020. [PMCID: PMC7153035 DOI: 10.1016/b978-0-12-816331-3.00008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Medical interventions to prevent and treat viral disease constitute evolutionary forces that may modify the genetic composition of viral populations that replicate in an infected host and influence the genomic composition of those viruses that are transmitted and progress at the epidemiological level. Given the adaptive potential of viruses in general and the RNA viruses in particular, the selection of viral mutants that display some degree of resistance to inhibitors or vaccines is a tangible challenge. Mutant selection may jeopardize control of the viral disease. Strategies intended to minimize vaccination and treatment failures are proposed and justified based on fundamental features of viral dynamics explained in the preceding chapters. The recommended use of complex, multiepitopic vaccines, and combination therapies as early as possible after initiation of infection falls under the general concept that complexity cannot be combated with simplicity. It also follows that sociopolitical action to interrupt virus replication and spread as soon as possible is as important as scientifically sound treatment designs to control viral disease on a global scale.
Collapse
|
116
|
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
|
117
|
Richardson D, Chan H, Bopage R, Lewis DA, Sawleshwarkar S, Chung C, Kok J. HIV-1 subtype variability and transmitted drug resistance in a culturally diverse population in Western Sydney, New South Wales, Australia. Sex Health 2020; 17:377-380. [PMID: 32687779 DOI: 10.1071/sh20013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/22/2020] [Indexed: 11/23/2022]
Abstract
Background Transmitted human immunodeficiency virus type 1 (HIV-1) drug resistance (TDR) is an important contributor to antiretroviral treatment failure, and is associated with HIV-1 transmission among men who have sex with men (MSM), non-MSM clusters and individuals diagnosed with concurrent sexually transmissible infections (STI). Western Sydney has a culturally diverse population, with a high proportion of non-Australian-born individuals. This study describes the prevalence of TDR and non-B HIV-1 subtypes in a clinic-based population. METHODS A clinic database was examined for all newly diagnosed treatment-naïve HIV-1 patients and information on their HIV-1 resistance and subtype, demographics including country of birth and diagnosis of a bacterial sexually transmissible infection was collected. RESULTS Data were available from 74/79 individuals (62 cis-male, 16 cis-female and 1 transgender woman). Of the 74 genotypes, the prevalence of non-B subtypes and TDR was 43/74 (58%; 95%CI = 46.9-69.3) and 14/74 (19%; 95%CI = 10.0 to 27.8). It was also found that 30/79 (38%) had a concurrent bacterial STI. TDR was associated with subtype B infection (OR 3.53; 95%CI = 1.41-8.82; P = 0.007) and being born in Australia (OR 12.0; 95%CI = 2.45-58.86; P = 0.002). CONCLUSION The relative prevalence of non-B HIV-1 subtypes and TDR is higher in Western Sydney than in the rest of Australia. TDR is associated with subtype B HIV-1 and being Australian born, suggesting ongoing local transmission. This highlights the diversity of the HIV epidemic locally and the need for interventions to prevent ongoing HIV transmission.
Collapse
Affiliation(s)
- Daniel Richardson
- Western Sydney Sexual Health Centre (Western Sydney Local Health District), Level 1, 162 Marsden Street, Parramatta, NSW 2150, Australia; and Corresponding author.
| | - Hubert Chan
- Western Sydney Sexual Health Centre (Western Sydney Local Health District), Level 1, 162 Marsden Street, Parramatta, NSW 2150, Australia
| | - Rohan Bopage
- Western Sydney Sexual Health Centre (Western Sydney Local Health District), Level 1, 162 Marsden Street, Parramatta, NSW 2150, Australia
| | - David A Lewis
- Western Sydney Sexual Health Centre (Western Sydney Local Health District), Level 1, 162 Marsden Street, Parramatta, NSW 2150, Australia; and Marie Bashir Institute for Infectious Diseases and Biosecurity and Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145, Australia
| | - Shailendra Sawleshwarkar
- Western Sydney Sexual Health Centre (Western Sydney Local Health District), Level 1, 162 Marsden Street, Parramatta, NSW 2150, Australia; and Marie Bashir Institute for Infectious Diseases and Biosecurity and Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145, Australia
| | - Charles Chung
- Western Sydney Sexual Health Centre (Western Sydney Local Health District), Level 1, 162 Marsden Street, Parramatta, NSW 2150, Australia
| | - Jen Kok
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute for Clinical Pathology and Medical Research, NSW Health Pathology, Westmead, NSW 2145, Australia
| |
Collapse
|
118
|
Bavaro DF, Di Carlo D, Rossetti B, Bruzzone B, Vicenti I, Pontali E, Zoncada A, Lombardi F, Di Giambenedetto S, Borghi V, Pecorari M, Milini P, Meraviglia P, Monno L, Saracino A. Pretreatment HIV drug resistance and treatment failure in non-Italian HIV-1-infected patients enrolled in ARCA. Antivir Ther 2020; 25:61-71. [PMID: 32118584 DOI: 10.3851/imp3349] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND An increase in pretreatment drug resistance (PDR) to first-line antiretroviral therapy (ART) in low-income countries has been recently described. Herein we analyse the prevalence of PDR and risk of virological failure (VF) over time among migrants to Italy enrolled in ARCA. METHODS HIV-1 sequences from ART-naive patients of non-Italian nationality were retrieved from ARCA database from 1998 to 2017. PDR was defined by at least one mutation from the reference 2009 WHO surveillance list. RESULTS Protease/reverse transcriptase sequences from 1,155 patients, mainly migrants from sub-Saharan Africa (SSA; 42%), followed by Latin America (LA; 25%) and Western countries (WE; 21%), were included. PDR was detected in 8.6% of sequences (13.1% versus 5.8% for B and non-B strains, respectively; P<0.001). 2.1% of patients carried a PDR for protease inhibitors (PIs; 2.1% versus 2.3%; P=0.893), 3.9% for nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs; 6.8% versus 2.1%; P<0.001) and 4.3% for non-nucleoside/nucleotide reverse transcriptase inhibitors (NNRTIs; 6.3% versus 3.1%; P=0.013). Overall, prevalence of PDR over the years remained stable, while it decreased for PIs in LA (P=0.021) and for NRTIs (P=0.020) among migrants from WE. Having more than one class of PDR (P=0.015 versus absence of PDR), higher viral load at diagnosis (P=0.008) and being migrants from SSA (P=0.001 versus WE) were predictive of VF, while a recent calendar year of diagnosis (P<0.001) was protective for VF. CONCLUSIONS PDR appeared to be stable over the years in migrants to Italy enrolled in ARCA; however, it still remains an important cause of VF together with viral load at diagnosis.
Collapse
Affiliation(s)
- Davide Fiore Bavaro
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari Medical School, Bari, Italy.,These authors equally contributed to this work
| | - Domenico Di Carlo
- Pediatric Clinical Research Center 'Romeo and Enrica Invernizzi', University of Milan, Milan, Italy.,These authors equally contributed to this work
| | - Barbara Rossetti
- Infectious Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Ilaria Vicenti
- Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | | | | | - Francesca Lombardi
- Università Cattolica del Sacro Cuore, Roma Italia, Istituto di Clinica Malattie Infettive, Rome, Italy
| | - Simona Di Giambenedetto
- Università Cattolica del Sacro Cuore, Roma Italia, Istituto di Clinica Malattie Infettive, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma Italia, UOC malattie infettive, Rome, Italy
| | - Vanni Borghi
- Clinica Malattie infettive, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Monica Pecorari
- SSD Virologia, Azienda Ospedaliero-Univeristaria Policlinico Modena, Modena, Italy
| | - Paola Milini
- Infectious Diseases Unit, Macerata Hospital, Macerata, Italy
| | - Paola Meraviglia
- 1st Division of Infectious Diseases, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Laura Monno
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari Medical School, Bari, Italy
| | - Annalisa Saracino
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, University of Bari Medical School, Bari, Italy
| |
Collapse
|
119
|
Ellis KE, Nawas GT, Chan C, York L, Fisher J, Connick E, Zangeneh TT. Clinical Outcomes Following the Use of Archived Proviral HIV-1 DNA Genotype to Guide Antiretroviral Therapy Adjustment. Open Forum Infect Dis 2019; 7:ofz533. [PMID: 31915714 PMCID: PMC6942490 DOI: 10.1093/ofid/ofz533] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/12/2019] [Indexed: 01/07/2023] Open
Abstract
Background Evidence regarding the safety of using proviral HIV-1 DNA genotype (DNA GT) to guide antiretroviral therapy (ART) is limited. We hypothesized that HIV RNA would not increase following ART adjustment guided by DNA GT in a university HIV clinic. Methods Data were obtained from electronic medical records of adult persons living with HIV-1 (PWH) who underwent DNA GT testing and changed ART between October 2014 and November 2017. Logistic regression was used to evaluate the effect of ART switch on HIV RNA over time. Results Eighty-three PWH had DNA GT performed, 66 (80%) switched ART, and 59 had postswitch follow-up. Data were analyzed pre-/postswitch for these 59 PWH (median age, 54 years; 71% LWH ≥10 years; 46% ≥2 previous regimens; 36% recent low-level viremia; 34% unknown medication history). On DNA GT, 58% had ≥1-class ART resistance, 34% ≥2-class, and 10% 3-class. Median follow-up (range) was 337 (34–647) days. There was no change in probability of HIV RNA ≥50 copies/mL over time (P > .05). At baseline, 76% had HIV RNA <50 vs 88% at last postswitch follow-up (P = .092). Protease inhibitor use decreased from 58% to 24% (P < .001). Average daily pills and dosing frequency decreased from 3.48 to 2.05 (P < .001) and 1.39 to 1.09 (P < .001), respectively; ART cost did not change. Conclusions DNA GT facilitated changes in ART in a treatment-experienced population without increases in HIV RNA. Decreased pill burden occurred without increased ART cost. Further studies to identify optimal use of DNA GT are needed.
Collapse
Affiliation(s)
- Kristen E Ellis
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA.,Department of Pharmacy Practice and Science, University of Arizona College of Pharmacy, Tucson, Arizona, USA
| | - George T Nawas
- Division of Clinical and Administrative Sciences, Xavier University of Louisiana College of Pharmacy, New Orleans, Louisiana, USA
| | - Connie Chan
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA.,Department of Pharmacy Practice and Science, University of Arizona College of Pharmacy, Tucson, Arizona, USA
| | - Lawrence York
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Julia Fisher
- Statistics Consulting Laboratory, BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| | - Elizabeth Connick
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Tirdad T Zangeneh
- Division of Infectious Diseases, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA
| |
Collapse
|
120
|
Noguera-Julian M. HIV drug resistance testing - The quest for Point-of-Care. EBioMedicine 2019; 50:11-12. [PMID: 31810819 PMCID: PMC6921291 DOI: 10.1016/j.ebiom.2019.11.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 01/14/2023] Open
Affiliation(s)
- Marc Noguera-Julian
- IrsiCaixa AIDS Research Institute, Badalona, Spain; University of Vic - Central University of Catalonia, Vic, Spain.
| |
Collapse
|
121
|
Raymond S, Nicot F, Abravanel F, Minier L, Carcenac R, Lefebvre C, Harter A, Martin-Blondel G, Delobel P, Izopet J. Performance evaluation of the Vela Dx Sentosa next-generation sequencing system for HIV-1 DNA genotypic resistance. J Clin Virol 2019; 122:104229. [PMID: 31809945 DOI: 10.1016/j.jcv.2019.104229] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 11/20/2022]
Abstract
BACKGROUND Patients on antiretroviral therapy could benefit from HIV-1 DNA resistance genotyping for exploring virological failure with low viral load or to guide treatment simplification. Few new generation sequencing data are available. OBJECTIVE To check that the automated deep sequencing Sentosa platform (Vela DX) detected minority resistant variants well enough for HIV DNA genotyping. STUDY DESIGN We evaluated the Sentosa SQ HIV genotyping assay with automated extraction on 40 DNA longitudinal samples from treatment-experienced patients by comparison with Sanger sequencing. HIV drug resistance was interpreted using the ANRS algorithm (v29) at the threshold of 20 % and 3 %. RESULTS The Sentosa SQ HIV genotyping assay was 100 % successful to amplify and sequence PR and RT and 86 % to amplify and sequence IN when the HIV DNA load was >2.5 log copies/million cells. The Sentosa and Sanger sequencing were concordant for predicting PR-RT resistance at the threshold of 20 % in 14/18 samples successfully sequenced. A higher level of resistance was predicted by Sentosa in three samples and by Sanger in one sample. The prevalence of resistance was 7 % to PI, 59 % to NRTI, 31 % to NNRTI and 20 % to integrase inhibitors using the Sentosa SQ genotyping assay at the threshold of 3 %. Seven additional mutations <20 % were detected using the Sentosa assay. CONCLUSION Automated DNA extraction and sequencing using the Sentosa SQ HIV genotyping assay accurately predicted HIV DNA drug resistance by comparison with Sanger. Prospective studies are needed to evaluate the clinical interest of HIV DNA genotyping.
Collapse
Affiliation(s)
- Stéphanie Raymond
- INSERM U1043, CNRS UMR 5282, Toulouse University Paul Sabatier, CPTP, Toulouse, F-31300 France; CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France.
| | - Florence Nicot
- CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France
| | - Florence Abravanel
- INSERM U1043, CNRS UMR 5282, Toulouse University Paul Sabatier, CPTP, Toulouse, F-31300 France; CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France
| | - Luce Minier
- CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France
| | - Romain Carcenac
- CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France
| | - Caroline Lefebvre
- CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France
| | - Agnès Harter
- CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France
| | - Guillaume Martin-Blondel
- INSERM U1043, CNRS UMR 5282, Toulouse University Paul Sabatier, CPTP, Toulouse, F-31300 France; CHU de Toulouse, Hôpital Purpan, Service des Maladies Infectieuses et Tropicales, Toulouse, F-31300 France
| | - Pierre Delobel
- INSERM U1043, CNRS UMR 5282, Toulouse University Paul Sabatier, CPTP, Toulouse, F-31300 France; CHU de Toulouse, Hôpital Purpan, Service des Maladies Infectieuses et Tropicales, Toulouse, F-31300 France
| | - Jacques Izopet
- INSERM U1043, CNRS UMR 5282, Toulouse University Paul Sabatier, CPTP, Toulouse, F-31300 France; CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, F-31300 France
| |
Collapse
|
122
|
Analysis of HIV-1 diversity, primary drug resistance and transmission networks in Croatia. Sci Rep 2019; 9:17307. [PMID: 31754119 PMCID: PMC6872562 DOI: 10.1038/s41598-019-53520-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/31/2019] [Indexed: 01/23/2023] Open
Abstract
Molecular epidemiology of HIV-1 infection in treatment-naive HIV-1 infected persons from Croatia was investigated. We included 403 persons, representing 92.4% of all HIV-positive individuals entering clinical care in Croatia in 2014–2017. Overall prevalence of transmitted drug resistance (TDR) was estimated at 16.4%. Resistance to nucleoside reverse transcriptase inhibitors (NRTIs), non-nucleoside RTI (NNRTIs) and protease inhibitors (PIs) was found in 11.4%, 6.7% and 2.5% of persons, respectively. Triple-class resistance was determined in 2.2% of individuals. In addition, a single case (1.0%) of resistance to integrase strand-transfer inhibitors (InSTIs) was found. Deep sequencing was performed on 48 randomly selected samples and detected additional TDR mutations in 6 cases. Phylogenetic inference showed that 347/403 sequences (86.1%) were part of transmission clusters and identified forward transmission of resistance in Croatia, even that of triple-class resistance. The largest TDR cluster of 53 persons with T215S was estimated to originate in the year 1992. Our data show a continuing need for pre-treatment HIV resistance testing in Croatia. Even though a low prevalence of resistance to InSTI was observed, surveillance of TDR to InSTI should be continued.
Collapse
|
123
|
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
|
124
|
Lack of HIV-1 integrase inhibitor resistance among 392 antiretroviral-naïve individuals in a tertiary care hospital in Beijing, China. AIDS 2019; 33:1945-1947. [PMID: 31491787 DOI: 10.1097/qad.0000000000002282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
: Reports of resistance to integrase strand transfer inhibitors (INSTIs) are now not uncommon. We analyzed the HIV int gene from plasma of antiretroviral-naïve individuals during acute and chronic HIV-1 infection. No individual with major INSTI mutations was identified. Two individuals harbored INSTI accessory mutations E157Q/T97A were detected for the first time. Our results emphasize the need to consider testing for INSTI resistance at baseline as this class of drugs is increasingly used in clinical routine.
Collapse
|
125
|
Rusere LN, Lockbaum GJ, Lee SK, Henes M, Kosovrasti K, Spielvogel E, Nalivaika EA, Swanstrom R, Yilmaz NK, Schiffer CA, Ali A. HIV-1 Protease Inhibitors Incorporating Stereochemically Defined P2' Ligands To Optimize Hydrogen Bonding in the Substrate Envelope. J Med Chem 2019; 62:8062-8079. [PMID: 31386368 PMCID: PMC6941148 DOI: 10.1021/acs.jmedchem.9b00838] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A structure-guided design strategy was used to improve the resistance profile of HIV-1 protease inhibitors by optimizing hydrogen bonding and van der Waals interactions with the protease while staying within the substrate envelope. Stereoisomers of 4-(1-hydroxyethyl)benzene and 4-(1,2-dihydroxyethyl)benzene moieties were explored as P2' ligands providing pairs of diastereoisomers epimeric at P2', which exhibited distinct potency profiles depending on the configuration of the hydroxyl group and size of the P1' group. While compounds with the 4-(1-hydroxyethyl)benzene P2' moiety maintained excellent antiviral potency against a panel of multidrug-resistant HIV-1 strains, analogues with the polar 4-(1,2-dihydroxyethyl)benzene moiety were less potent, and only the (R)-epimer incorporating a larger 2-ethylbutyl P1' group showed improved potency. Crystal structures of protease-inhibitor complexes revealed strong hydrogen bonding interactions of both (R)- and (S)-stereoisomers of the hydroxyethyl group with Asp30'. Notably, the (R)-dihydroxyethyl group was involved in a unique pattern of direct hydrogen bonding interactions with the backbone amides of Asp29' and Asp30'. The SAR data and analysis of crystal structures provide insights for optimizing these promising HIV-1 protease inhibitors.
Collapse
Affiliation(s)
- Linah N. Rusere
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Gordon J. Lockbaum
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Sook-Kyung Lee
- Department of Biochemistry and Biophysics, and the UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Mina Henes
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Klajdi Kosovrasti
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Ean Spielvogel
- Department of Biochemistry and Biophysics, and the UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Ellen A. Nalivaika
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Ronald Swanstrom
- Department of Biochemistry and Biophysics, and the UNC Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Akbar Ali
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
126
|
Abstract
Approximately 20% of people with HIV in the United States prescribed antiretroviral therapy are not virally suppressed. Thus, optimal management of virologic failure has a critical role in the ability to improve viral suppression rates to improve long-term health outcomes for those infected and to achieve epidemic control. This article discusses the causes of virologic failure, the use of resistance testing to guide management after failure, interpretation and relevance of HIV drug resistance patterns, considerations for selection of second-line and salvage therapies, and management of virologic failure in special populations.
Collapse
Affiliation(s)
- Suzanne M McCluskey
- Division of Infectious Diseases, Harvard Medical School, Massachusetts General Hospital, 55 Fruit Street, GRJ5, Boston, MA 02114, USA.
| | - Mark J Siedner
- Division of Infectious Diseases, Harvard Medical School, Massachusetts General Hospital, 55 Fruit Street, GRJ5, Boston, MA 02114, USA
| | - Vincent C Marconi
- Division of Infectious Diseases, Department of Global Health, Emory University School of Medicine, Rollins School of Public Health, Health Sciences Research Building, 1760 Haygood Dr NE, Room W325, Atlanta, GA 30322, USA
| |
Collapse
|
127
|
Áy É, Müller V, Mezei M, Pocskay Á, Koroknai A, Müller D, Győri Z, Marschalkó M, Tóth B, Kárpáti S, Lakatos B, Szlávik J, Takács M, Minárovits J. Transmitted drug resistance in newly diagnosed and treatment-naïve HIV type 1-infected patients in Hungary. J Glob Antimicrob Resist 2019; 20:124-130. [PMID: 31330377 DOI: 10.1016/j.jgar.2019.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/28/2019] [Accepted: 07/11/2019] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES Transmitted human immunodeficiency virus type 1 (HIV-1) drug resistance (TDR) may affect the success of first-line antiretroviral treatment. This study aimed to monitor the presence of HIV-1 strains carrying transmitted drug resistance-associated mutations (TDRMs) in newly diagnosed and treatment-naïve patients in Hungary. METHODS This study included 168 HIV-infected individuals diagnosed between 2013-2017; most of them (93.5%) belonged to the homo/bisexual population. HIV-1 subtypes and TDRMs were determined by analysing the protease and reverse transcriptase coding regions of the pol gene by the Stanford HIV Drug Resistance Database. Transmission clusters among patients were identified using phylogenetic analysis. RESULTS Although subtype B HIV-1 strains were predominant (87.5%), non-B subtypes including F, A, CRF01_AE, CRF02_AG, D and G were also recorded, especially in young adults. The overall prevalence of TDR was 10.7% (18 of 168; 95% CI: 6.9-16.3%). Subtype B HIV-1 strains carried most of the TDRMs (94.4%). Nucleoside reverse transcriptase inhibitor (NRTI)-associated mutations were the most prevalent indicators of TDR (16 of 168; 9.5%; 95% CI: 5.9-14.9%), followed by mutations conferring resistance to non-nucleoside reverse transcriptase inhibitors (NNRTIs) (2 of 168; 1.2%; 95% CI: 0.3-4.2%) and protease inhibitors (PIs) (1 of 168, 0.6%; 95% CI: 0.1-3.3%). Phylogenetic analysis revealed that most NRTI-associated resistance mutations were associated with a single monophyletic clade, suggesting early single-source introduction and ongoing spread of this drug-resistant HIV-1 strain. CONCLUSIONS Onward transmission of drug-resistant subtype B HIV-1 strains accounted for the majority of TDRs observed among treatment-naïve HIV-infected individuals in Hungary.
Collapse
Affiliation(s)
- Éva Áy
- National Public Health Institute, Department of Retroviruses, National Reference Laboratory of HIV, Albert Flórián út 2-6, H-1097 Budapest, Hungary
| | - Viktor Müller
- Eötvös Loránd University, Institute of Biology, Department of Plant Systematics, Ecology and Theoretical Biology, Pázmány Péter sétány 1/C, H-1117 Budapest, Hungary
| | - Mária Mezei
- National Public Health Institute, Department of Retroviruses, National Reference Laboratory of HIV, Albert Flórián út 2-6, H-1097 Budapest, Hungary
| | - Ágnes Pocskay
- National Public Health Institute, Department of Retroviruses, National Reference Laboratory of HIV, Albert Flórián út 2-6, H-1097 Budapest, Hungary
| | - Anita Koroknai
- National Public Health Institute, Department of Retroviruses, National Reference Laboratory of HIV, Albert Flórián út 2-6, H-1097 Budapest, Hungary
| | - Dalma Müller
- Eötvös Loránd University, Institute of Biology, Department of Plant Systematics, Ecology and Theoretical Biology, Pázmány Péter sétány 1/C, H-1117 Budapest, Hungary
| | - Zoltán Győri
- National Public Health Institute, Department of Retroviruses, National Reference Laboratory of HIV, Albert Flórián út 2-6, H-1097 Budapest, Hungary
| | - Márta Marschalkó
- Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Mária utca 41, H-1085 Budapest, Hungary
| | - Béla Tóth
- Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Mária utca 41, H-1085 Budapest, Hungary
| | - Sarolta Kárpáti
- Semmelweis University, Department of Dermatology, Venereology and Dermatooncology, Mária utca 41, H-1085 Budapest, Hungary
| | - Botond Lakatos
- Center for HIV, National Institute of Hematology and Infectious Diseases, South-Pest Central Hospital, Albert Flórián út 5-7, H-1097 Budapest, Hungary
| | - János Szlávik
- Center for HIV, National Institute of Hematology and Infectious Diseases, South-Pest Central Hospital, Albert Flórián út 5-7, H-1097 Budapest, Hungary
| | - Mária Takács
- National Public Health Institute, Directorate for Clinical and Public Health Microbiology, Albert Flórián út 2-6, H-1097 Budapest, Hungary; Semmelweis University, Institute of Medical Microbiology, Nagyvárad tér 4, H-1089 Budapest, Hungary
| | - János Minárovits
- University of Szeged, Department of Oral Biology and Experimental Dental Research, Tisza Lajos krt. 64, H-6720 Szeged, Hungary.
| |
Collapse
|
128
|
Banin AN, Tuen M, Bimela JS, Tongo M, Zappile P, Khodadadi‐Jamayran A, Nanfack AJ, Okonko IO, Meli J, Wang X, Mbanya D, Ngogang J, Gorny MK, Heguy A, Fokunang C, Duerr R. Near full genome characterization of HIV-1 unique recombinant forms in Cameroon reveals dominant CRF02_AG and F2 recombination patterns. J Int AIDS Soc 2019; 22:e25362. [PMID: 31353798 PMCID: PMC6661401 DOI: 10.1002/jia2.25362] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 07/04/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION In Cameroon, a manifold diversity of HIV strains exists with CRF02_AG and unique recombinant forms (URFs) being the predominant strains. In recent years, a steady increase in URFs and clade F2 viruses has been monitored through partial genome sequencing. There is an information gap in the characterization of emerging URFs along the full genome, which is needed to address the challenges URFs pose towards diagnosis, treatment and HIV-1 vaccine design. METHOD Eighteen Cameroonian URFs from samples collected between the years 2000 and 2015 were studied using a newly developed near full genome sequencing (NFGS) protocol based on variable nested RT-PCRs with a versatile primer set. Near full genomes were characterized for recombination patterns and sequence signatures with possible impact on antiretroviral treatment or Env-directed immune responses. Third-generation sequencing (3GS) of near full or half genomes (HGs) gave insight into intra-patient URF diversity. RESULTS The characterized URFs were composed of a broad variety of subtypes and recombinants including A, F, G, CRF01_AE, CRF02_AG and CRF22_01A1. Phylogenetic analysis unveiled dominant CRF02_AG and F2 recombination patterns. 3GS indicated a high intra-patient URF diversity with up to four distinct viral sub-populations present in plasma at the same time. URF pol genomic analysis revealed a number of accessory drug resistance mutations (DRMs) in the ART-naïve participants. Genotypic env analysis suggests CCR5 usage in 14/18 samples and identified deviations at residues, critical for gp120/gp41 interphase and CD4 binding site broadly neutralizing antibodies in more than half of the studied URFs. V1V2 sites of immune pressure in the human RV144 vaccine study varied in more than a third of URFs. CONCLUSIONS This study identified novel mosaic patterns in URFs in Cameroon. In line with the regional predominance of CRF_02AG and the increased prevalence of clade F2, prominent CRF_02AG and F2 background patterns were observed underlying the URFs. In the context of the novel mosaic genomes, the impact of the identified accessory DRMs and Env epitope variations on treatment and immune control remains elusive. The evolving diversity of HIV-1 URFs in Cameroon requires continuous monitoring to respond to the increasing challenges for diagnosis, antiretroviral treatment and prevention.
Collapse
Affiliation(s)
- Andrew N Banin
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
- Faculty of Medicine and Biomedical SciencesUniversity of Yaoundé 1YaoundéCameroon
| | - Michael Tuen
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
| | - Jude S Bimela
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
- Faculty of ScienceDepartment of BiochemistryUniversity of Yaoundé 1YaoundéCameroon
| | - Marcel Tongo
- Center of Research for Emerging and Re‐Emerging Diseases (CREMER)Institute of Medical Research and Study of Medicinal PlantsYaoundéCameroon
| | - Paul Zappile
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
| | - Alireza Khodadadi‐Jamayran
- Applied Bioinformatics Laboratories (ABL) and Genome Technology Center (GTC)Division of Advanced Research Technologies (DART)New York University Langone Medical CenterNew YorkNYUSA
| | - Aubin J Nanfack
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
- Medical Diagnostic CenterYaoundéCameroon
- Chantal Biya International Reference Center for Research on HIV/AIDS Prevention and ManagementYaoundéCameroon
| | - Iheanyi O Okonko
- Virus Research UnitDepartment of MicrobiologyUniversity of Port HarcourtPort HarcourtNigeria
| | | | - Xiaohong Wang
- Manhattan Veterans Affairs Harbor Healthcare SystemsNew YorkNYUSA
| | - Dora Mbanya
- Faculty of Medicine and Biomedical SciencesUniversity of Yaoundé 1YaoundéCameroon
| | - Jeanne Ngogang
- Faculty of Medicine and Biomedical SciencesUniversity of Yaoundé 1YaoundéCameroon
| | - Miroslaw K Gorny
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
| | - Adriana Heguy
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
| | - Charles Fokunang
- Faculty of Medicine and Biomedical SciencesUniversity of Yaoundé 1YaoundéCameroon
| | - Ralf Duerr
- Department of PathologyNew York University School of MedicineNew YorkNYUSA
- Manhattan Veterans Affairs Harbor Healthcare SystemsNew YorkNYUSA
| |
Collapse
|
129
|
Takou D, Fokam J, Teto G, Santoro MM, Ceccherini-Silberstein F, Nanfack AJ, Sosso SM, Dambaya B, Salpini R, Billong SC, Gori C, Fokunang CN, Cappelli G, Colizzi V, Perno CF, Ndjolo A. HIV-1 drug resistance testing is essential for heavily-treated patients switching from first- to second-line regimens in resource-limited settings: evidence from routine clinical practice in Cameroon. BMC Infect Dis 2019; 19:246. [PMID: 30871487 PMCID: PMC6419466 DOI: 10.1186/s12879-019-3871-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 03/04/2019] [Indexed: 01/17/2023] Open
Abstract
Background With the phase-out of stavudine (d4T), change to first-line regimens with zidovudine (AZT) or tenofovir (TDF) in resource-limited settings (RLS) might increase risks of cross-resistance to nucleos(t) ide reverse transcriptase inhibitors (NRTI). This would restrict the scope of switching to the World Health Organisation (WHO)-recommended standard second-line combinations (SLC) without HIV drug resistance (HIVDR)-testing in routine clinical practice. Methods An observational study was conducted among 101 Cameroonian patients (55.4% male, median [IQR] age 34 [10–41] years) failing first-line antiretroviral therapy (ART) in 2016, and stratified into three groups according to NRTIs exposure: exposure to both thymidine analogues AZT “and” D4T (group-A, n = 55); exposure to both TDF and AZT “or” D4T (group-B, n = 22); exposure solely to D4T (group-C, n = 24). Protease-reverse transcriptase HIVDR was interpreted using the HIVdb penalty scores (≥60: high-resistance; 20–59: intermediate-resistance; < 20: susceptible). The acceptable threshold for potential-efficacy was set at 80%. Results The median [IQR] CD4, viral RNA, and time on ART, were respectively 129 [29–466] cells/μl, 71,630 [19,041-368,000] copies/ml, and 4 [2–5] years. Overall HIVDR-level was 89.11% (90/101), with 83.2% harbouring M184 V (high-level 3TC/FTC-resistance) and only 1.98% (2/101) major HIVDR-mutations to ritonavir-boosted protease-inhibitors (PI/r). Thymidine-analogue mutations (TAMs)-1 [T215FY (46.53%), M41 L (22.77%), L210 W (8.91%)], with cross-resistance to AZT and TDF, were higher compared to TAMs-2 [D67N (21.78%), K70R (19.80%), K219QE (18.81%)]. As expected, K65R was related with TDF-exposure: 0% (0/55) in group-A, 22.72% (5/22) group-B, 4.17% (1/24) group-C (p = 0.0013). The potential-efficacy of AZT vs. TDF was respectively 43.64% (24/55) vs. 70.91% (39/55) in group-A (p = 0.0038); 63.64% (14/22) vs. 68.28% (15/22) in group-B (p = 1.0000); and 37.50% (9/24) vs. 83.33% (20/24) in group-C (p = 0.0032). CRF02_AG was the prevailing subtype (63.40%), followed by CRF11.cpx (8.91%), A1 (7.92%), G (5.94%); without any significant effect of the subtype-distribution on HIVDR (92.2% in CRF02_AG vs. 83.8% in non-AG; p = 0.204). Conclusion First-line ART-failure exhibits high-level NRTI-resistance, with potential lower-efficacy of AZT compared to TDF. Significantly, using our 80% efficacy-threshold, only patients without NRTI-substitution on first-line could effectively switch to SLC following the WHO-approach. Patients with multiple NRTI-substitutions (exposed to both thymidine-analogues and TDF) on first-line ART would require HIVDR-testing to select active NRTIs for SLC. Electronic supplementary material The online version of this article (10.1186/s12879-019-3871-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Desire Takou
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon
| | - Joseph Fokam
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon. .,Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon. .,National HIV Drug Resistance prevention and surveillance Working Group, Ministry of Public Health, Yaoundé, Cameroon.
| | - Georges Teto
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon
| | | | | | - Aubin Joseph Nanfack
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon
| | - Samuel Martin Sosso
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon
| | - Béatrice Dambaya
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon
| | | | - Serge Clotaire Billong
- National HIV Drug Resistance prevention and surveillance Working Group, Ministry of Public Health, Yaoundé, Cameroon.,Surveillance, Research, Planning, Monitoring and Evaluation service, Central Technical Group, National AIDS Control Committee, Yaounde, Cameroon
| | - Caterina Gori
- National Institute of Infectious Diseases Lazzaro Spallanzani, Rome, Italy
| | | | - Giulia Cappelli
- Institute of Cellular Biology and Neurobiology (IBCN), Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Vittorio Colizzi
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon.,University of Rome Tor Vergata, Rome, Italy
| | - Carlo-Federico Perno
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon.,University of Rome Tor Vergata, Rome, Italy.,University of Milan, Milan, Italy
| | - Alexis Ndjolo
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management, Yaoundé, Cameroon.,Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| |
Collapse
|
130
|
Alvarez M, Casas P, de Salazar A, Chueca N, Guerrero-Beltran C, Rodríguez C, Imaz A, Espinosa N, García-Bujalance S, Pérez-Elías MJ, García-Alvarez M, Iribarren JA, Santos J, Dalmau D, Aguilera A, Vinuesa D, Gutiérrez F, Piérola B, Molina JM, Peraire J, Portilla I, Gómez-Sirvent JL, Olalla J, Galera C, Blanco JR, Riera M, García-Fraile L, Navarro G, Curran A, Poveda E, García F, Moreno S, Jarrín I, Dalmau D, Navarro ML, González MI, Blanco JL, Garcia F, Rubio R, Iribarren JA, Gutiérrez F, Vidal F, Berenguer J, González J, Alejos B, Hernando V, Moreno C, Iniesta C, Sousa LMG, Perez NS, Muñoz-Fernández MÁ, García-Merino IM, Fernández IC, Rico CG, de la Fuente JG, Concejo PP. Surveillance of transmitted drug resistance to integrase inhibitors in Spain: implications for clinical practice. J Antimicrob Chemother 2019; 74:1693-1700. [DOI: 10.1093/jac/dkz067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/17/2019] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Marta Alvarez
- Unidad de Microbiología Clínica, Hospital Universitario San Cecilio, Granada, Instituto de Investigacion Ibs., Granada, Spain
| | - Paz Casas
- Unidad de Microbiología Clínica, Hospital Universitario San Cecilio, Granada, Instituto de Investigacion Ibs., Granada, Spain
| | - Adolfo de Salazar
- Unidad de Microbiología Clínica, Hospital Universitario San Cecilio, Granada, Instituto de Investigacion Ibs., Granada, Spain
| | - Natalia Chueca
- Unidad de Microbiología Clínica, Hospital Universitario San Cecilio, Granada, Instituto de Investigacion Ibs., Granada, Spain
| | - Carlos Guerrero-Beltran
- Unidad de Microbiología Clínica, Hospital Universitario San Cecilio, Granada, Instituto de Investigacion Ibs., Granada, Spain
| | | | - Arkaitz Imaz
- Unidad de VIH e ITS, Departamento de Enfermedades Infecciosas, Hospital Universitari de Bellvitge-IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Nuria Espinosa
- Unidad de Enfermedades Infecciosas, Hospital Virgen del Rocio, Sevilla, Spain
| | | | | | - Mónica García-Alvarez
- Unidad de Microbiología Clínica, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Jose Antonio Iribarren
- Unidad de Enfermedades Infecciosas, Hospital Universitario Donostia, Instituto BioDonostia, Donostia, Spain
| | - Jesús Santos
- Unidad de Enfermedades Infecciosas, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - David Dalmau
- Unidad de Enfermedades Infecciosas, Hospital Universitario Mutua Terrasa, Terrasa, Spain
| | - Antonio Aguilera
- Servicio y Departamento de Microbiología, Complejo Hospitalario Universitario de Santiago y Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - David Vinuesa
- Unidad de Enfermedades Infecciosas, Hospital Universitario, Universitario San Cecilio, Granada, Spain
| | - Félix Gutiérrez
- Unidad de Enfermedades Infecciosas, Hospital Universitario de Elche & Universidad Miguel Hernández, Alicante, Spain
| | - Beatriz Piérola
- Unidad de Enfermedades Infecciosas, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - José Miguel Molina
- Unidad de Microbiología Clínica, Hospital Universitario La Fe, Valencia, Spain
| | - Joaquim Peraire
- Unidad de Enfermedades Infecciosas, Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Irene Portilla
- Unidad de Enfermedades Infecciosas, Hospital Universitario Alicante, Alicante, Spain
| | - Juan Luis Gómez-Sirvent
- Unidad de Enfermedades Infecciosas, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Julián Olalla
- Unidad de Enfermedades Infecciosas, Hospital Costa del Sol, Marbella, Spain
| | - Carlos Galera
- Unidad de Enfermedades Infecciosas, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - José Ramón Blanco
- Unidad de Enfermedades Infecciosas, Hospital Universitario San Pedro, Logroño, Spain
| | - Melchor Riera
- Unidad de Enfermedades Infecciosas, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Lucio García-Fraile
- Unidad de Enfermedades Infecciosas, Hospital Universitario La Princesa, Madrid, Spain
| | - Gemma Navarro
- Unidad de Enfermedades Infecciosas, Hospital Universitario Parc Taulí, Sabadell, Spain
| | - Adrían Curran
- Servicio de Enfermedades Infecciosas, Hospital Universitario Vall d´Hebron, Barcelona, Spain
| | - Eva Poveda
- Group of Virology and Pathogenesis, Galicia Sur Health Research Institute (IIS Galicia Sur)-Complexo Hospitalario Universitario de Vigo, SERGAS-UVigo, Spain
| | - Federico García
- Unidad de Microbiología Clínica, Hospital Universitario San Cecilio, Granada, Instituto de Investigacion Ibs., Granada, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Kamelian K, Lepik KJ, Chau W, Yip B, Zhang WW, Lima VD, Robbins MA, Woods C, Olmstead A, Joy JB, Barrios R, Harrigan PR. Prevalence of Human Immunodeficiency Virus-1 Integrase Strand Transfer Inhibitor Resistance in British Columbia, Canada Between 2009 and 2016: A Longitudinal Analysis. Open Forum Infect Dis 2019; 6:ofz060. [PMID: 30895202 PMCID: PMC6419991 DOI: 10.1093/ofid/ofz060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/06/2019] [Indexed: 11/12/2022] Open
Abstract
Background Integrase strand transfer inhibitors (INSTIs) are highly efficacious and well tolerated antiretrovirals with fewer adverse side-effects relative to other classes of antiretrovirals. The use of INSTIs raltegravir, elvitegravir, and dolutegravir has increased dramatically over recent years. However, there is limited information about the evolution and prevalence of INSTI resistance mutations in clinical human immunodeficiency virus populations. Methods Human immunodeficiency virus-1-positive individuals ≥19 years were included if they received ≥1 dispensed prescription of antiretroviral therapy (ART) in British Columbia between 2009 and 2016 (N = 9358). Physician-ordered drug resistance tests were analyzed and protease inhibitor (PI), reverse-transcriptase inhibitor (RT), and INSTI resistance were defined as having ≥1 sample with a combined, cumulative score ≥30 by Stanford HIV Drug Resistance Algorithm version 7.0.1. Results Although most ART-treated individuals were tested for PI and RT resistance, INSTI resistance testing lagged behind the uptake of INSTIs among INSTI-treated individuals (11% in 2009; 34% in 2016). The prevalence of INSTI resistance was relatively low, but it increased from 1 to 7 per 1000 ART-treated individuals between 2009 and 2016 (P < .0001, R2 = 0.98). Integrase strand transfer inhibitor resistance mutations increased at integrase codons 66, 97, 140, 148, 155, and 263. Conclusions The prevalence of INSTI resistance remains low compared with PI and RT resistance in ART-treated populations but is expanding with increased INSTI use.
Collapse
Affiliation(s)
- Kimia Kamelian
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada.,University of British Columbia, Department of Medicine, Division of AIDS, Vancouver, Canada
| | - Katherine J Lepik
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada.,Pharmacy Department, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - William Chau
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Benita Yip
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Wendy W Zhang
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada.,University of British Columbia, Department of Medicine, Division of AIDS, Vancouver, Canada
| | - Viviane Dias Lima
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada.,University of British Columbia, Department of Medicine, Division of AIDS, Vancouver, Canada
| | - Marjorie A Robbins
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Conan Woods
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada
| | - Andrea Olmstead
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada.,University of British Columbia, Department of Medicine, Division of AIDS, Vancouver, Canada
| | - Jeffrey B Joy
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada.,University of British Columbia, Department of Medicine, Division of AIDS, Vancouver, Canada
| | - Rolando Barrios
- BC Centre for Excellence in HIV/AIDS, Vancouver, British Columbia, Canada.,University of British Columbia, School of Population and Public Health, Vancouver, Canada
| | - P Richard Harrigan
- University of British Columbia, Department of Medicine, Division of AIDS, Vancouver, Canada
| |
Collapse
|
132
|
Prevalence of HIV-1 transmitted drug resistance and viral suppression among recently diagnosed adults in São Paulo, Brazil. Arch Virol 2018; 164:699-706. [PMID: 30569276 DOI: 10.1007/s00705-018-04122-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/15/2018] [Indexed: 10/27/2022]
Abstract
HIV-1 transmitted drug resistance (TDR) mutations may reduce the efficacy of antiretroviral therapy (ART), but pre-treatment testing to determine the virus genotype can improve the efficacy of ART. Unfortunately, issues related to cost and logistics of pre-treatment testing limit its use in resource-limited settings. We studied 596 ART-naive individuals who were newly diagnosed from 2014 to 2016 in São Paulo, Brazil, to evaluate TDR and virological outcome after 48 weeks of genotype-guided therapy. One or more TDR (based on the WHO surveillance list) was observed in 10.9% (CI 95%, 8.6-13.6) of the sequences, the most common of which was the K103 N mutation, which confers resistance to first-generation drugs of the non-nucleoside reverse transcriptase inhibitor (NNRTI) antiretroviral drug class. Dual-class (1%, 6/596) and triple-class (0.34%, 2/596) resistance were uncommon. After 48 weeks of treatment with ART, infection was suppressed to below 200 copies/mL in most patients (95%), with full suppression (RNA target not detected) in 65%. The following characteristics at patient enrollment were independently associated with a lack of full suppression: CD4 T cell counts below 500 cells/µL, viremia above 100,000 copies/mL, older age, and TDR to NNRTI. The rates of resistance were intermediate, but genotype-guided therapy resulted in high rates of viral suppression. The observed resistance profile should not be an obstacle to the use of the dolutegravir-based regimen now recommended in Brazil, but genotype testing may be warranted before initiating first-generation NNRTI-based regimens.
Collapse
|
133
|
Limited Marginal Utility of Deep Sequencing for HIV Drug Resistance Testing in the Age of Integrase Inhibitors. J Clin Microbiol 2018; 56:JCM.01443-18. [PMID: 30305383 PMCID: PMC6258839 DOI: 10.1128/jcm.01443-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/02/2018] [Indexed: 02/02/2023] Open
Abstract
HIV drug resistance genotyping is a critical tool in the clinical management of HIV infections. Although resistance genotyping has traditionally been conducted using Sanger sequencing, next-generation sequencing (NGS) is emerging as a powerful tool due to its ability to detect low-frequency alleles. HIV drug resistance genotyping is a critical tool in the clinical management of HIV infections. Although resistance genotyping has traditionally been conducted using Sanger sequencing, next-generation sequencing (NGS) is emerging as a powerful tool due to its ability to detect low-frequency alleles. However, the clinical value added from NGS approaches to antiviral resistance testing remains to be demonstrated. We compared the variant detection capacity of NGS versus Sanger sequencing methods for resistance genotyping in 144 drug resistance tests (105 protease-reverse transcriptase tests and 39 integrase tests) submitted to our clinical virology laboratory over a four-month period in 2016 for Sanger-based HIV drug resistance testing. NGS detected all true high-frequency drug resistance mutations (>20% frequency) found by Sanger sequencing, with greater accuracy in one instance of a Sanger-detected false positive. Freely available online NGS variant callers HyDRA and PASeq were superior to Sanger methods for interpretations of allele linkage and automated variant calling. NGS additionally detected low-frequency mutations (1 to 20% frequency) associated with higher levels of drug resistance in 30/105 (29%) protease-reverse transcriptase tests and 4/39 (10%) integrase tests. In clinical follow-up of 69 individuals for a median of 674 days, we did not find a difference in rates of virological failure between individuals with and without low-frequency mutations, although rates of virological failure were higher for individuals with drug-relevant low-frequency mutations. However, all 27 individuals who experienced virological failure reported poor adherence to their drug regimen during the preceding follow-up time, and all 19 who subsequently improved their adherence achieved viral suppression at later time points, consistent with a lack of clinical resistance. In conclusion, in a population with low antiviral resistance emergence, NGS methods detected numerous instances of minor alleles that did not result in subsequent bona fide virological failure due to antiviral resistance.
Collapse
|
134
|
Silver N, Paynter M, McAllister G, Atchley M, Sayir C, Short J, Winner D, Alouani DJ, Sharkey FH, Bergefall K, Templeton K, Carrington D, Quiñones-Mateu ME. Characterization of minority HIV-1 drug resistant variants in the United Kingdom following the verification of a deep sequencing-based HIV-1 genotyping and tropism assay. AIDS Res Ther 2018; 15:18. [PMID: 30409215 PMCID: PMC6223033 DOI: 10.1186/s12981-018-0206-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/30/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The widespread global access to antiretroviral drugs has led to considerable reductions in morbidity and mortality but, unfortunately, the risk of virologic failure increases with the emergence, and potential transmission, of drug resistant viruses. Detecting and quantifying HIV-1 drug resistance has therefore become the standard of care when designing new antiretroviral regimens. The sensitivity of Sanger sequencing-based HIV-1 genotypic assays is limited by its inability to identify minority members of the quasispecies, i.e., it only detects variants present above ~ 20% of the viral population, thus, failing to detect minority variants below this threshold. It is clear that deep sequencing-based HIV-1 genotyping assays are an important step change towards accurately monitoring HIV-infected individuals. METHODS We implemented and verified a clinically validated HIV-1 genotyping assay based on deep sequencing (DEEPGEN™) in two clinical laboratories in the United Kingdom: St. George's University Hospitals Healthcare NHS Foundation Trust (London) and at NHS Lothian (Edinburgh), to characterize minority HIV-1 variants in 109 plasma samples from ART-naïve or -experienced individuals. RESULTS Although subtype B HIV-1 strains were highly prevalent (44%, 48/109), most individuals were infected with non-B subtype viruses (i.e., A1, A2, C, D, F1, G, CRF02_AG, and CRF01_AE). DEEPGEN™ was able to accurately detect drug resistance-associated mutations not identified using standard Sanger sequencing-based tests, which correlated significantly with patient's antiretroviral treatment histories. A higher proportion of minority PI-, NRTI-, and NNRTI-resistance mutations was detected in NHS Lothian patients compared to individuals from St. George's, mainly M46I/L and I50 V (associated with PIs), D67 N, K65R, L74I, M184 V/I, and K219Q (NRTIs), and L100I (NNRTIs). Interestingly, we observed an inverse correlation between intra-patient HIV-1 diversity and CD4+ T cell counts in the NHS Lothian patients. CONCLUSIONS This is the first study evaluating the transition, training, and implementation of DEEPGEN™ between three clinical laboratories in two different countries. More importantly, we were able to characterize the HIV-1 drug resistance profile (including minority variants), coreceptor tropism, subtyping, and intra-patient viral diversity in patients from the United Kingdom, providing a rigorous foundation for basing clinical decisions on highly sensitive and cost-effective deep sequencing-based HIV-1 genotyping assays in the country.
Collapse
|
135
|
McGee KS, Okeke NL, Hurt CB, McKellar MS. Canary in the Coal Mine? Transmitted Mutations Conferring Resistance to All Integrase Strand Transfer Inhibitors in a Treatment-Naive Patient. Open Forum Infect Dis 2018; 5:ofy294. [PMID: 30515433 PMCID: PMC6262110 DOI: 10.1093/ofid/ofy294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/02/2018] [Indexed: 12/28/2022] Open
Abstract
Transmitted drug resistance to the integrase strand transfer inhibitor (INSTI) class of antiretrovirals is very rare. We present a case of a treatment-naive female patient with human immunodeficiency virus harboring resistance to all INSTIs, including bictegravir and dolutegravir.
Collapse
Affiliation(s)
- Kara S McGee
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | - Nwora Lance Okeke
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | - Christopher B Hurt
- Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill
| | - Mehri S McKellar
- Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|