1
|
Dähne T, Jaki L, Gosert R, Fuchs J, Krumbholz A, Nägele K, Pletz MW, Khanna N, Leuzinger K, Panning M. Herpes Simplex Virus and Drug Resistance - Comprehensive Update on Resistance Mutations and Implications for Clinical Management: A Narrative Review. Clin Microbiol Infect 2025:S1198-743X(25)00237-X. [PMID: 40349973 DOI: 10.1016/j.cmi.2025.04.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/04/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Antiviral drug resistance in herpes simplex virus 1 and 2 (HSV-1 & 2) is a significant clinical challenge, particularly in immunocompromised patients. Drug susceptibility testing (DST) aids clinical management and can be conducted through genotypic (partial genome sequencing) or phenotypic (cell culture) methods. Both have inherent limitations: genotypic DST is limited by outdated datasets lacking information on new helicase-primase inhibitors and corresponding phenotypic data as well as sparse clinical correlations. Phenotypic DST is mainly hampered by a lack of standardization and timely results. OBJECTIVES To compile an up-to-date and comprehensive HSV drug resistance dataset encompassing all reported drug resistance-associated mutations (DRMs), polymorphisms, and viral phenotypes. To aggregate clinical conditions with available DST data. SOURCES A PubMed search identified studies (January 2016-September 2024) on DRMs associated with resistance to aciclovir, penciclovir, brivudine, foscarnet, cidofovir, amenamevir, and pritelivir. Data from a previous HSV resistance dataset (pre-2016) were also included. CONTENT In this review, we summarize novel mutations in the thymidine kinase, polymerase, and helicase-primase gene of HSV conferring resistance to antiviral drugs. Clinical information was available for 513 mutations. In 90% of these (461 cases), viral phenotype and clinical assessment were congruent. However, 10% of cases not responding to antiviral therapy showed phenotypically susceptible virus isolates. We present a framework for clinical and diagnostic management of cases with drug resistant HSV infection. IMPLICATIONS This dataset paves the way to harmonize reporting of DRMs for diagnostic labs and to accelerate genotypic DST interpretation through aggregated data. Ongoing large-scale data collection of genotypic, phenotypic, and clinical data is crucial for evidence-based management of HSV antiviral resistance and clinical guidelines.
Collapse
Affiliation(s)
- Theo Dähne
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Consulting Laboratory for HSV and VZV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lena Jaki
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rainer Gosert
- Clinical Virology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Jonas Fuchs
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andi Krumbholz
- Institute for Infection Medicine, Christian-Albrechts-Universität zu Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany; Labor Dr. Krause und Kollegen Medizinisches Versorgungszentrum GmbH, Kiel, Germany
| | - Klaudia Nägele
- Clinical Virology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Mathias W Pletz
- Institute of Infectious Diseases and Infection Control and Center for Sepsis Care and Control, Jena University Hospital, Friedrich-Schiller-University Jena, Jena, Germany
| | - Nina Khanna
- Division of Infectious Diseases, University and University Hospital Basel, Basel, Switzerland
| | - Karoline Leuzinger
- Clinical Virology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Marcus Panning
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Consulting Laboratory for HSV and VZV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
2
|
Biswas A, Choudhuri I, Huang K, Sun Q, Sali A, Echeverria I, Haldane A, Levy RM, Lyumkis D. Evolutionary Sequence and Structural Basis for the Epistatic Origins of Drug Resistance in HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.30.651576. [PMID: 40364913 PMCID: PMC12073831 DOI: 10.1101/2025.04.30.651576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The emergence of drug resistance in the human immunodeficiency virus (HIV) remains a formidable challenge to the long-term efficacy of antiretroviral therapy (ART). A growing body of evidence highlights the critical role of epistasis, the dependence of mutational effects on the sequence context, in shaping the fitness landscape of HIV under ART-induced selection pressure. However, the biophysical origins of the epistatic interactions involved in engendering drug-resistance mutations (DRMs) remain unclear. Are the mutational correlations "intrinsic" to the properties of the protein, or do they arise because of drug binding? We use a Potts sequence-covariation statistical energy model built on patient-derived HIV-1 protein sequences to construct computational double mutant cycles that probe pairwise epistasis for all observed mutations across the three major HIV drug-target enzymes. We find that the strongest epistatic effects occur between mutations at residue positions that frequently mutate during the course of ART, termed resistance-associated positions. To investigate the structural origins of the strongest epistatic interactions, we perform ∼100 free energy perturbation molecular dynamics simulations, revealing that the primary contribution to the pairwise epistasis between DRMs arises from cooperative effects on protein stability and folding as an intrinsic consequence of the protein mutational landscape. The results collectively reinforce a mechanism of resistance evolution whereby viruses escape drug pressure by selectively engendering mutations at "intrinsically" coupled sites, allowing them to cooperatively ameliorate fitness detriments incurred by individual DRMs. Significance Epistasis refers to the phenomenon where the effect of a mutation on protein structure and function is dependent on the genetic sequence background of the mutation, resulting in the combined effect of mutations being non-additive. Epistasis plays a significant role in the evolution of drug resistance in viruses such as HIV under therapeutic selection pressure. We combine a protein sequence coevolutionary model and molecular dynamics free energy simulations to identify and probe the mechanistic origins of the strongest epistatic interactions connecting HIV drug-resistance mutations. The work establishes a foundation to probe the molecular bases of epistasis and predict the evolution of resistance predicated on the knowledge of epistatic interaction networks.
Collapse
|
3
|
Omoding D, Musinguzi N, Boum Y, Muzoora C, Kigozi S, Hunt PW, Martin JN, Bangsberg DR, Haberer JE, Siedner MJ, McCluskey SM, Lee GQ. Subtypes A1 and D, and recombinant HIV-1 natural polymorphisms associated with lenacapavir drug resistance in Uganda. J Antimicrob Chemother 2025; 80:955-961. [PMID: 39881519 PMCID: PMC11962371 DOI: 10.1093/jac/dkaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Lenacapavir, a novel HIV-1 capsid inhibitor, shows promise for treating MDR HIV-1, as well as for pre-exposure prophylaxis (PrEP) in prevention of HIV infection. Its unique mechanism and lack of cross-resistance with other antiretroviral classes make lenacapavir a significant addition to HIV therapy. The clinical trials CALIBRATE and CAPELLA have demonstrated high viral suppression rates in both ART-naive individuals and individuals with MDR HIV-1. Lenacapavir-associated resistance mutations, such as M66I and Q67H, rarely seen as natural polymorphisms in lenacapavir-naive populations, are predominantly studied in subtype B HIV-1. OBJECTIVES Our study aimed to investigate the prevalence of lenacapavir resistance-associated mutations in HIV-1 subtypes A1 and D in a cohort of individuals living with HIV-1 from southwestern Uganda. METHODS Utilizing plasma samples from ART-naive adults living in Uganda, HIV-1 Gag p24 (capsid) sequences were analysed for lenacapavir resistance mutations. RESULTS Among 546 lenacapavir-naive participants, no major lenacapavir resistance-associated mutations were found. Minor mutations were present in 1.6% of participants, with T107A being the most common. Longitudinal data indicated the persistence of T107A for at least 3 years post-ART initiation in one participant. Phylogenetic analysis indicated individuals carrying T107A were found independently in distinct locations within the tree, suggesting that T107A might have arisen from multiple distinct base substitution events. Shannon entropy analysis showed high variability in certain capsid sites, but none overlapped with known lenacapavir resistance sites. CONCLUSIONS These findings suggest a low prevalence of naturally occurring lenacapavir resistance mutations in Uganda, supporting lenacapavir's potential efficacy in this region.
Collapse
Affiliation(s)
- Daniel Omoding
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Nicholas Musinguzi
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Yap Boum
- Institut Pasteur de Bangui, Bangui, Central African Republic
| | - Conrad Muzoora
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Simone Kigozi
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Peter W Hunt
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jeffrey N Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | | | - Jessica E Haberer
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mark J Siedner
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Suzanne M McCluskey
- Department of Community Health, Mbarara University of Science and Technology, Mbarara, Uganda
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Guinevere Q Lee
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
4
|
Koul M, Kaushik S, Singh K, Sharma D. VITALdb: to select the best viroinformatics tools for a desired virus or application. Brief Bioinform 2025; 26:bbaf084. [PMID: 40063348 PMCID: PMC11892104 DOI: 10.1093/bib/bbaf084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/14/2025] [Accepted: 02/17/2025] [Indexed: 05/13/2025] Open
Abstract
The recent pandemics of viral diseases, COVID-19/mpox (humans) and lumpy skin disease (cattle), have kept us glued to viral research. These pandemics along with the recent human metapneumovirus outbreak have exposed the urgency for early diagnosis of viral infections, vaccine development, and discovery of novel antiviral drugs and therapeutics. To support this, there is an armamentarium of virus-specific computational tools that are currently available. VITALdb (VIroinformatics Tools and ALgorithms database) is a resource of ~360 viroinformatics tools encompassing all major viruses (SARS-CoV-2, influenza virus, human immunodeficiency virus, papillomavirus, herpes simplex virus, hepatitis virus, dengue virus, Ebola virus, Zika virus, etc.) and several diverse applications [structural and functional annotation, antiviral peptides development, subspecies characterization, recognition of viral recombination, inhibitors identification, phylogenetic analysis, virus-host prediction, viral metagenomics, detection of mutation(s), primer designing, etc.]. Resources, tools, and other utilities mentioned in this article will not only facilitate further developments in the realm of viroinformatics but also provide tremendous fillip to translate fundamental knowledge into applied research. Most importantly, VITALdb is an inevitable tool for selecting the best tool(s) to carry out a desired task and hence will prove to be a vital database (VITALdb) for the scientific community. Database URL: https://compbio.iitr.ac.in/vitaldb.
Collapse
Affiliation(s)
- Mira Koul
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Shalini Kaushik
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Kavya Singh
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Deepak Sharma
- Computational Biology and Translational Bioinformatics (CBTB) Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|
5
|
Hung TM, Bang LVN, Van Duyet L. Development of HIV Drug-Resistance Mutations and Antiretroviral Efficacy Among Vietnamese Patients After Failure of 5-Year First-Line Therapy. J Clin Lab Anal 2025; 39:e25157. [PMID: 39907180 PMCID: PMC11904817 DOI: 10.1002/jcla.25157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND The emergence of drug-resistant mutations in human immunodeficiency virus (HIV) over time presents a challenge to treatment. We describe the development of drug-resistance mutations and ART efficacy reduction in Vietnamese patients with failure of first-line ART during a 5-year period. METHODS This is a 5-year observational cohort study with HIV viral loads of patients evaluated annually for 5 years (2017-2022) at the hospitals in Vietnam. Patients with a viral load ≥ 1000 copies/mL were subjected to identifying mutations in reverse transcriptase, protease, and integrase to evaluate HIV resistance and the efficacy of ART. RESULTS After 5 years of monitoring the HIV load of 2932 patients on ART, 75 (2.56%) patients had concurrent virological failure at all 5 years. In 2017, only 2/75 HIV strains possessed Protease Inhibitor (PI) resistance mutations, while 75/75 HIV strains had both Nucleoside Reverse Transcriptase Inhibitors (NRTIs) and Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs) resistance mutations. Only four PI resistance variants were found, while 40 and 32 mutations were resistant to NRTIs and NNRTIs. After 5 years, the number of HIV PI resistance mutations had increased to 14, with 13 new mutations emerging. There were six novel mutations associated with resistance to NRTIs, NNRTIs, and the proportion of preexisting mutations increased from 1.3% to 13.3%. Furthermore, HIV sensitivity to ART decreased from 2.7% to 18.6%. CONCLUSION After 5 years, HIV had increased resistance mutations to PIs, NRTIs, and NNRTIs, with PI resistance mutations increasing the most rapidly, and the decrease in HIV sensitivity to PIs was higher than that to NRTIs and NNRTIs.
Collapse
Affiliation(s)
- Than Manh Hung
- Emergency DepartmentNational Hospital for Tropical DiseasesHanoiVietnam
- Infectious DepartmentUniversity of Medicine and Pharmacy, Vietnam National UniversityHanoiVietnam
| | | | - Le Van Duyet
- Micobiology and Moclecular Biology DepartmentNational Hospital for Tropical DiseasesHanoiVietnam
| |
Collapse
|
6
|
Lule DB, Ssemwanga D, Kaleebu P, Tully DC. The utility of integrating nanopore sequencing into routine HIV-1 drug resistance surveillance. Microb Genom 2025; 11:001375. [PMID: 40111248 PMCID: PMC11925199 DOI: 10.1099/mgen.0.001375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/01/2025] [Indexed: 03/22/2025] Open
Abstract
HIV continues to be a significant global public health concern. In 2022, an estimated 29.8 million people living with HIV received antiretroviral treatment (ART). From this, an estimated 10-15% of individuals living with HIV have drug-resistant strains of the virus. Testing for resistance to antiretroviral drugs is recommended before initiating ART. However, such services are often inaccessible due to costs and the need for complex laboratory infrastructure. The assessment of HIV drug resistance (HIVDR) relies on genotyping sequencing and algorithms to interpret genotypic resistance test results. Genotypic assays involve Sanger sequencing of the reverse transcriptase (RT), protease (PR) and integrase (IN) genes of circulating RNA in plasma to detect mutations that are known to confer drug resistance. While state-of-the-art sequencing technologies have swept the globe and enhanced our global pandemic response capabilities, they are still sparingly used for HIVDR surveillance. The scale-up of ART, especially in low- and middle-income countries, necessitates the establishment of cheap, expeditious and decentralized methods for HIVDR monitoring. Here, we outline how one low-capital next-generation sequencing platform, namely, nanopore sequencing, could augment efforts in expanding HIVDR surveillance efforts, especially in resource-limited settings. We discuss that because of its versatility, nanopore sequencing can accelerate HIVDR surveillance in conjunction with scaling up ART efforts and outline some of the challenges that need to be considered before its widespread and routine adaptation to detect drug resistance rapidly.
Collapse
Affiliation(s)
- Daniel Bugembe Lule
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
- Medical Research Council/Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine Uganda Unit, Plot 51-59 Nakiwogo Road, P. O. Box 49, Entebbe, Uganda
- St. Georges University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK
| | - Deogratius Ssemwanga
- Medical Research Council/Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine Uganda Unit, Plot 51-59 Nakiwogo Road, P. O. Box 49, Entebbe, Uganda
- Uganda Virus Research Institute, Plot 51-59 Nakiwogo Road, P. O. Box 49, Entebbe, Uganda
| | - Pontiano Kaleebu
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
- Medical Research Council/Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine Uganda Unit, Plot 51-59 Nakiwogo Road, P. O. Box 49, Entebbe, Uganda
- Uganda Virus Research Institute, Plot 51-59 Nakiwogo Road, P. O. Box 49, Entebbe, Uganda
| | - Damien C. Tully
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
- Medical Research Council/Uganda Virus Research Institute & London School of Hygiene and Tropical Medicine Uganda Unit, Plot 51-59 Nakiwogo Road, P. O. Box 49, Entebbe, Uganda
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
7
|
Mahmud R, Krullaars Z, van Osch J, Rickett D, Brumme ZL, Hensley KS, Rokx C, Gruters RA, van Kampen JJA, Mesplède T. Computational and Population-Based HLA Permissiveness to HIV Drug Resistance-Associated Mutations. Pathogens 2025; 14:207. [PMID: 40137693 PMCID: PMC11944876 DOI: 10.3390/pathogens14030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
The presentation of HIV peptides by the human leukocyte antigen (HLA) complex to CD8+ cytotoxic T-cells (CTLs) is critical to limit viral pathogenesis. HIV can mutate to evade HLA-restricted CTL responses and resist antiretroviral drugs, raising questions about how it balances these evolutionary pressures. Here, we used a computational approach to assess how drug resistance-associated mutations (RAMs) affect the binding of HIV-1 subtype B or C peptides to the most prevalent HLA alleles in US, European, and South African populations. We predict RAMs that may be favored in certain populations and report the under-representation of Y181C in people expressing HLA-B*57:01. This finding agreed with our computational predictions when Y181C was at the major anchor site P2, suggesting the potential relevance of our approach. Overall, our findings lay out a conceptual framework to study the implications of HLA alleles on the emergence of HIV RAMs at the individual and population levels.
Collapse
Affiliation(s)
- Rizwan Mahmud
- Viroscience Department, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (R.M.); (Z.K.); (J.v.O.); (R.A.G.); (J.J.A.v.K.)
| | - Zoë Krullaars
- Viroscience Department, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (R.M.); (Z.K.); (J.v.O.); (R.A.G.); (J.J.A.v.K.)
| | - Jolieke van Osch
- Viroscience Department, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (R.M.); (Z.K.); (J.v.O.); (R.A.G.); (J.J.A.v.K.)
| | - David Rickett
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, BC V6Z1Y6, Canada; (D.R.); (Z.L.B.)
| | - Zabrina L. Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, BC V6Z1Y6, Canada; (D.R.); (Z.L.B.)
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A1S6, Canada
| | - Kathryn S. Hensley
- Departments of Internal Medicine (Infectious Diseases) and Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (K.S.H.); (C.R.)
| | - Casper Rokx
- Departments of Internal Medicine (Infectious Diseases) and Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (K.S.H.); (C.R.)
| | - Rob A. Gruters
- Viroscience Department, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (R.M.); (Z.K.); (J.v.O.); (R.A.G.); (J.J.A.v.K.)
| | - Jeroen J. A. van Kampen
- Viroscience Department, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (R.M.); (Z.K.); (J.v.O.); (R.A.G.); (J.J.A.v.K.)
| | - Thibault Mesplède
- Viroscience Department, Erasmus University Medical Center, 3015GD Rotterdam, The Netherlands; (R.M.); (Z.K.); (J.v.O.); (R.A.G.); (J.J.A.v.K.)
| |
Collapse
|
8
|
Fraysse J, Priest J, Turner M, Hill S, Jones B, Verdier G, Letang E. Real-World Effectiveness and Tolerability of Dolutegravir and Lamivudine 2-Drug Regimen in People Living with HIV: Systematic Literature Review and Meta-Analysis. Infect Dis Ther 2025; 14:357-383. [PMID: 39826080 PMCID: PMC11829890 DOI: 10.1007/s40121-024-01103-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/20/2025] Open
Abstract
INTRODUCTION Dolutegravir (DTG) + lamivudine (3TC) demonstrated high rates of virologic suppression (VS) and low rates of virologic failure (VF), discontinuation, and drug resistance in randomized trials. Real-world evidence can support treatment effectiveness, safety, and tolerability in clinical practice and aid in treatment decisions. METHODS A systematic literature review (SLR) was conducted to identify studies using DTG + 3TC (January 2013-March 2024). Studies were screened to include observational studies reporting 48- or 96-week on-treatment VS, VF, or discontinuation outcomes; proportions of individuals with each outcome at each time point were estimated using random- and common-effects models. RESULTS Of 249 SLR-identified publications, 43 reported consistently defined outcomes of interest at comparable time points, representing 1480 individuals naive to antiretroviral therapy (ART) and 12,234 individuals with prior ART experience. At weeks 48 and 96, respectively, estimated proportions (95% CIs; random-effects model) with on-treatment VS were high (naive to ART, 0.964 [0.945-0.979] and 0.902 [0.816-0.966]; prior ART, 0.966 [0.950-0.980] and 0.971 [0.946-0.990]), with low estimated proportions experiencing VF (naive to ART, 0.001 [0.000-0.013] and 0.001 [0.000-0.008]; prior ART, 0.009 [0.005-0.015] and 0.015 [0.007-0.024]) and discontinuations for any reason (naive to ART, 0.052 [0.019-0.097] and 0.130 [0.084-0.183]; prior ART, 0.067 [0.042-0.098] and 0.084 [0.047-0.130]). Across identified studies (> 44,000 unique individuals), those reporting resistance outcomes at VF/blip (regardless of emergence) detected integrase strand transfer inhibitor (INSTI) mutations in 0 of 2346 individuals naive to ART and 0.02% (4/20,060) of individuals with prior ART experience (S147G, R263K, G118R + E138K, T66A + G118R + E138K); additionally, N155H was reported in an individual using DTG + 3TC with unknown baseline ART status. CONCLUSION Overall treatment outcomes in real-world settings confirm the efficacy, tolerability, and high barrier to resistance seen in phase 3 trials across diverse populations, including those naive to ART or with prior ART experience.
Collapse
Affiliation(s)
| | - Julie Priest
- ViiV Healthcare, 406 Blackwell Street, Suite 300, Durham, NC, 27701, USA.
| | | | | | | | | | | |
Collapse
|
9
|
Novitsky V, Steingrimsson J, Guang A, Dunn CW, Howison M, Gillani FS, Hague J, Fulton J, Bertrand T, Bhattarai L, MacAskill M, Bandy U, Hogan J, Kantor R. Dynamics of clustering rates in the Rhode Island HIV-1 epidemic. AIDS 2025; 39:105-114. [PMID: 39527774 PMCID: PMC11717628 DOI: 10.1097/qad.0000000000004062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Characterizing HIV clustering rates and their trends over time can improve understanding a local epidemic and enhance its control. METHODS Leveraging an academic-public health partnership in Rhode Island, we explored longitudinal dynamics of statewide clustering rates among key populations from 1991 to 2023. Partial HIV-1 pol sequences were grouped by year of HIV-1 diagnosis. Molecular clusters were identified in cumulative annual phylogenies. Overall clustering rates, and clustering rates of newly diagnosed and prevalent infections, and of specific sociodemographic characteristics of key populations over time were determined. Mann-Kendall statistics were used to estimate clustering rate trends and relationships among groups. RESULTS By the end of 2023, 2630 individuals with sequences represented the statewide epidemic in Rhode Island. Overall clustering rates increased from 7% in 1991 to 46% in 2023, correlating with cumulative sequence increase. Clustering rates of newly diagnosed and prevalent infections significantly increased over time, higher in newly diagnosed individuals since the early 2000s. Increases were also observed among groups defined by gender, age, transmission risks, race, mental illness, HIV-1 subtypes, and country of birth, with some crossovers and divergence patterns over time. CONCLUSION Exploring dynamics of HIV clustering rates over three decades in a statewide HIV-1 epidemic expanded its characterization and provided insight into its evolving changes. These dynamics may indicate a gradual shift towards a more concentrated and localized HIV-1 epidemic, highlighting important opportunities for targeted interventions to effectively prevent new HIV transmissions.
Collapse
Affiliation(s)
| | | | | | | | - Mark Howison
- Research Improving People’s Lives, Providence, RI, USA
| | | | | | | | | | | | | | - Utpala Bandy
- Rhode Island Department of Health, Providence, RI, USA
| | | | | |
Collapse
|
10
|
Bikinesi L, Spinelli MA, Nyoni N, Mouton D, Mengistu A, Kamangu J, Konstantinus I, Kalimugogo P, Mutandi G, Negussie F, Wang G, Welty S, McFarland W, Beard RS, Haberer J, McCluskey S, Gandhi M, Hong SY. The impact of adherence counseling incorporating a point of care urine tenofovir assay on virologic suppression among individuals failing tenofovir-lamivudine-dolutegravir: A pre-post intervention study. Int J Infect Dis 2025; 151:107328. [PMID: 39647577 PMCID: PMC11748914 DOI: 10.1016/j.ijid.2024.107328] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/11/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
OBJECTIVES To examine if point-of-care (POC) urine tenofovir testing-informed counseling could be used to improve virologic suppression (VS) among participants with virologic failure (VF) after ≥1 prior round of enhanced adherence counseling (EAC). METHODS Participants were enrolled from 42 clinics across Namibia. At each monthly medication pick-up, participants completed the POC urine test and received EAC informed by this testing (EAC+). If VS was not achieved after 3 months of EAC+, up to 3 additional rounds of EAC+ were provided, with resistance testing at month (M)9. RESULTS Of 310 potentially eligible participants across 42 clinics in Namibia, we enrolled 211 participants with VF (median age 33 years, 61% female); 195 reached M3 defined as receiving EAC+ and follow-up viral load testing; 169 achieved VS within M3 (87%, P < 0.001) and 97% by M9 (181/186) compared to 40% (22/55) prior to the intervention (P < 0.001). Resistance testing was performed in five remaining participants with VF at M9, of whom 1/5 (20%) developed dolutegravir resistance. CONCLUSION The urine tenofovir assay when incorporated into adherence counseling has potential to be a cost-effective intervention among participants failing tenofovir-based regimens, increasing VS to 97% in those failing Tenofovir-Lamivudine-Dolutegravir. Encouraging results of this pre-post intervention will be rigorously tested in a randomized trial.
Collapse
Affiliation(s)
| | - Matthew A Spinelli
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco (UCSF), USA
| | - Ntombizodwa Nyoni
- University of California San Francisco Global Programs for Research and Training, Windhoek, Namibia.
| | | | | | | | | | - Pearl Kalimugogo
- Centers for Disease Control and Prevention, Division of Global HIV and Tuberculosis (DGHT), Windhoek, Namibia
| | - Gram Mutandi
- Centers for Disease Control and Prevention, Division of Global HIV and Tuberculosis, Atlanta, Georgia, USA
| | - Fekir Negussie
- University of California San Francisco Global Programs for Research and Training, Windhoek, Namibia
| | - Guohong Wang
- Abbott Toxicology Unit, Abbott Pharmaceuticals, USA
| | - Susie Welty
- Department of Epidemiology and Biostatistics, UCSF, USA
| | | | - R Suzanne Beard
- Centers for Disease Control and Prevention, Division of Global HIV and Tuberculosis, Atlanta, Georgia, USA
| | - Jessica Haberer
- Harvard Medical School and Massachusetts General Hospital, MA, USA
| | | | - Monica Gandhi
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco (UCSF), USA
| | - Steven Y Hong
- Centers for Disease Control and Prevention, Division of Global HIV and Tuberculosis, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Naidu D, Oduro-Kwateng E, Soliman MES, Ndlovu SI, Mkhwanazi NP. Alternaria alternata (Fr) Keissl Crude Extract Inhibits HIV Subtypes and Integrase Drug-Resistant Strains at Different Stages of HIV Replication. Pharmaceuticals (Basel) 2025; 18:189. [PMID: 40006004 PMCID: PMC11859181 DOI: 10.3390/ph18020189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The development of HIV drug resistance to current antiretrovirals, and the antiretrovirals' inability to cure HIV, provides the need of developing novel drugs that inhibit HIV-1 subtypes and drug-resistance strains. Fungal endophytes, including Alternaria alternata, stand out for their potentially antiviral secondary metabolites. Hence, this study investigates the anti-HIV activities and mechanism of action of the A. alternata crude extract against different HIV-1 subtypes and integrase-resistant mutant strains. Methods: Cytotoxicity of the A. alternata crude extract on TZM-bl cells using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay was performed. The crude extract antiviral activity against subtypes A, B, C, and D and integrase drug-resistant strain T66K and S230R was determined using a luciferase-based antiviral assay. Luciferase and p24 ELISA-based time-of-addition assays were used to determine the mechanism of action of the crude extract. Docking scores and protein ligand interactions of integrase T66K and S230R strains against the identified bioactive compounds were determined. Results: The crude extract CC50 was 300 μg/mL and not cytotoxic to the TZM-bl cell lines. In HIV-1 subtypes A, B, C, and D, the crude extract exhibited 100% inhibition and therapeutic potential. The A. alternata crude extract had strong anti-HIV-1 activity against integrase strand transfer drug-resistant strains T66K and S230R, with a 0.7265- and 0. 8751-fold increase in susceptibility. The crude extract had antiviral activity during attachment, reverse transcription, integration, and proteolysis. In silico calculations showed compounds 2,3-2H-Benzofuran-2-one, 3,3,4,6-tetramethyl-, 3-Methyl-1,4-diazabicyclo[4.3.0]nonan-2,5-dione, N-acetyl, Coumarin, 3,4-dihydro-4,5,7-trimethyl-, Cyclopropanecarboxamide, N-cycloheptyl, Pyrrolo[1,2-a]pyrazine-1,4-dione, and hexahydro-3-(2-methylpropyl)- crude extract bioactive compounds had strong docking scores and diverse binding mechanisms with integrase. Conclusions: The A. alternata crude extract demonstrates strong antiviral activity against different HIV-1 subtypes and integrase drug-resistance strains. The extract inhibited various stages of the HIV-1 life cycle. The bioactive compounds 2,3-2H-Benzofuran-2-one, 3,3,4,6-tetramethyl-, 3-Methyl-1,4-diazabicyclo[4.3.0]nonan-2,5-dione, N-acetyl, Coumarin, 3,4-dihydro-4,5,7-trimethyl-, Cyclopropanecarboxamide, N-cycloheptyl, Pyrrolo[1,2-a]pyrazine-1,4-dione, and hexahydro-3-(2-methylpropyl)- may be responsible for the antiviral activity of A. alternata.
Collapse
Affiliation(s)
- Darian Naidu
- HIV Pathogenesis Programme, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu-Natal, Durban 4041, South Africa;
| | - Ernest Oduro-Kwateng
- Molecular Bio-Computation and Drug Design Research Group, School of Health Sciences, University of KwaZulu Natal, Durban 4041, South Africa (M.E.S.S.)
| | - Mahmoud E. S. Soliman
- Molecular Bio-Computation and Drug Design Research Group, School of Health Sciences, University of KwaZulu Natal, Durban 4041, South Africa (M.E.S.S.)
| | - Sizwe I. Ndlovu
- Unit for Environmental Sciences and Management, North-West University, Potchefstroom 2520, South Africa;
| | - Nompumelelo P. Mkhwanazi
- HIV Pathogenesis Programme, School of Laboratory Medicine and Medical Science, College of Health Science, University of KwaZulu-Natal, Durban 4041, South Africa;
| |
Collapse
|
12
|
Glyschewski L, Hahn A, Rohde H, Lütgehetmann M, Feldt T, Sarfo FS, Phillips RO, Dompreh A, Asibey SO, Boateng R, Weinreich F, Frickmann H, Eberhardt KA. Replicative co-infections with human immunodeficiency virus 1 and 2 as well as hepatitis B and C virus in Ghanaian individuals. Eur J Microbiol Immunol (Bp) 2024; 14:346-360. [PMID: 39475752 PMCID: PMC11836648 DOI: 10.1556/1886.2024.00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/14/2024] [Indexed: 12/19/2024] Open
Abstract
Background The study assessed replicative human immunodeficiency virus-(HIV-) infection and replicative co-infections as well as molecular determinants of reduced susceptibility towards anti-retroviral therapy in a Ghanaian population of known HIV patients and a control group. Methods Real-time PCRs for HIV-1, HIV-2, hepatitis B virus (HBV) and hepatitis C virus (HCV) were run with serum samples from known Ghanaian HIV-patients (n = 975) and control individuals (n = 105). For 108 individuals, HIV-sequence analysis was performed. Results Prevalence of replicative HIV-1 infection was 59.8% (583/975) in the known HIV-positive population and 2.9% (3/105) in the controls. Prevalences of replicative HBV-infection were comparable with 3.4% (33/975) in the HIV-positive individuals and 3.8% (4/105) in the controls. HIV-2 and HCV sequences were not recorded. Almost perfect concordance between two compared HIV-1-PCR assays was indicated by Fleiss' Kappa >0.8. Sanger sequencing indicated CRF_02AG, G and A3 as the quantitatively dominating HIV-1 subtypes, a minority of 3.4% CXCR4 tropism and high detection rates of mutations mediating reduced susceptibility towards nucleoside reverse transcriptase inhibitors (71.9%, 64/89), non-nucleoside reverse transcriptase inhibitors (95.5%, 85/89), protease inhibitors (95.9%, 93/97) and integrase inhibitors (22.4%, 22/98). Conclusions The assessment did not suggest HIV-triggered increased replication of HBV and HCV in the investigated Ghanaian population.
Collapse
Affiliation(s)
- Lynn Glyschewski
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, Hamburg, Germany
| | - Andreas Hahn
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Torsten Feldt
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Medical Center Düsseldorf, Düsseldorf, Germany
| | - Fred Stephen Sarfo
- Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Medicine, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | | | - Albert Dompreh
- Department of Clinical Microbiology, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | | | - Richard Boateng
- Department of Clinical Microbiology, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | | | - Hagen Frickmann
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, Hamburg, Germany
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Kirsten Alexandra Eberhardt
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Department of Medicine, University Medical Center, Hamburg, Germany
| |
Collapse
|
13
|
Byun H, Papathanasopoulos MA, Steegen K, Basson AE. Thymidine Analogue Mutations with M184V Significantly Decrease Phenotypic Susceptibility of HIV-1 Subtype C Reverse Transcriptase to Islatravir. Viruses 2024; 16:1888. [PMID: 39772195 PMCID: PMC11680407 DOI: 10.3390/v16121888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 01/30/2025] Open
Abstract
Islatravir (ISL) is the first-in-class nucleoside reverse transcriptase translocation inhibitor (NRTtI) with novel modes of action. Data on ISL resistance are currently limited, particularly to HIV-1 non-B subtypes. This study aimed to assess prevalent nucleos(t)ide reverse transcriptase inhibitor (NRTI)-resistant mutations in HIV-1 subtype C for their phenotypic resistance to ISL. Prevalent single and combinations of NRTI-resistant mutations were selected from a routine HIV-1 genotypic drug resistance testing database and introduced into HIV-1 subtype C-like pseudoviruses, which were then tested for ISL susceptibility. Single NRTI-resistant mutations were susceptible or showed only a low level of resistance to ISL. This included thymidine analogue mutations (TAMs, i.e., M41L, D67N, K70R, T215FY, and K219EQ) and non-TAMs (i.e., A62V, K65R, K70ET, L74IV, A114S, Y115F, and M184V). Combinations of M184V with one or more additional NRTI-resistant mutations generally displayed reduced ISL susceptibilities. This was more prominent for combinations that included M184V+TAMs, and particularly M184V+TAM-2 mutations. Combinations that included M184V+K65R did not impact significantly on ISL susceptibility. Our study suggests that ISL would be effective in treating people living with HIV (PLWH) failing tenofovir disoproxil fumarate (TDF)/lamivudine (3TC) or TDF/emtricitabine (FTC)-containing regimens, but would be less effective in PLH failing zidovudine (AZT) with 3TC or FTC-containing regimens.
Collapse
Affiliation(s)
- Hyeonah Byun
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (H.B.); (M.A.P.)
| | - Maria Antonia Papathanasopoulos
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (H.B.); (M.A.P.)
| | - Kim Steegen
- National Priority Programme, National Health Laboratory Service, Johannesburg 2192, South Africa;
- Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Adriaan Erasmus Basson
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (H.B.); (M.A.P.)
| |
Collapse
|
14
|
Tilloy V, Díaz-González D, Laplace L, Bisserier E, Chou S, Rawlinson WD, Boivin G, Baldanti F, Lazzarotto T, Andrei G, Hirsch HH, Marcos MÁ, Michel D, Hantz S, Alain S. Comprehensive Herpesviruses Antiviral drug Resistance Mutation Database (CHARMD). Antiviral Res 2024; 231:106016. [PMID: 39349222 DOI: 10.1016/j.antiviral.2024.106016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
A comprehensive and accessible Herpesvirus drug resistance database was designed to serve as an international reference for diagnosis and clinical studies. This database available at https://www.unilim.fr/cnr-herpesvirus/outils/codexmv/includes both resistance-related mutations and natural polymorphisms. Initially designed for human cytomegalovirus, it will be expanded to include herpes simplex and varicella-zoster viruses. Newly published mutations and new mutations reported by users or collaborating expert laboratories will be reviewed by an international committee of reference laboratories before inclusion in the database. Coupled with the Herpesvirus Sequence Analysis tool (HSA) mutation reports from NGS or Sanger sequences, it will be an open source for researchers in the field of Herpesviruses. We hope to fill this unmet need for the development and standardization of resistance genotyping.
Collapse
Affiliation(s)
- Valentin Tilloy
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; UF9481 Bioinformatique, CHU Limoges, Limoges, France; UF8843 Génomique médicale, CHU Limoges, Limoges, France; Inserm, U1092, RESINFIT, Université de Limoges, Limoges, France.
| | - Daniel Díaz-González
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; UF8843 Génomique médicale, CHU Limoges, Limoges, France
| | - Lisa Laplace
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France
| | - Emilien Bisserier
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France
| | - Sunwen Chou
- Department of Veterans Affairs Medical Center, Portland. Oregon, USA
| | - William D Rawlinson
- Virology and OTDS Laboratories (SAViD), Prince of Wales Hospital, Barker Street, Randwick, NSW, 2031, NSW Health Pathology Randwick, Australia
| | - Guy Boivin
- Centre de recherche en infectiologie, CHU de Québec-Université Laval, Canada
| | | | - Tiziana Lazzarotto
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Graciela Andrei
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hans H Hirsch
- Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - María Ángeles Marcos
- Servicio de Microbiología, Hospital Clinic, ISGlobal (Instituto de Salud Global de Barcelona), Barcelona, Spain
| | | | - Sébastien Hantz
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; Inserm, U1092, RESINFIT, Université de Limoges, Limoges, France
| | - Sophie Alain
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; UF8843 Génomique médicale, CHU Limoges, Limoges, France; Inserm, U1092, RESINFIT, Université de Limoges, Limoges, France; FHU SUPORT, CHU Limoges, Limoges, France.
| |
Collapse
|
15
|
Martin MA, Reynolds SJ, Foley BT, Nalugoda F, Quinn TC, Kemp SA, Nakalanzi M, Kankaka EN, Kigozi G, Ssekubugu R, Gupta RK, Abeler-Dörner L, Kagaayi J, Ratmann O, Fraser C, Galiwango RM, Bonsall D, Grabowski MK. Population dynamics of HIV drug resistance during treatment scale-up in Uganda: a population-based longitudinal study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.14.23297021. [PMID: 39417110 PMCID: PMC11482865 DOI: 10.1101/2023.10.14.23297021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Background Clinical studies have reported rising pre-treatment HIV drug resistance during antiretroviral treatment (ART) scale-up in Africa, but representative data are limited. We estimated population-level drug resistance trends during ART expansion in Uganda. Methods We analyzed data from the population-based open Rakai Community Cohort Study conducted at agrarian, trading, and fishing communities in southern Uganda between 2012 and 2019. Consenting participants aged 15-49 were HIV tested and completed questionnaires. Persons living with HIV (PLHIV) provided samples for viral load quantification and virus deep-sequencing. Sequence data were used to predict resistance. Population prevalence of class-specific resistance and resistance-conferring substitutions were estimated using robust log-Poisson regression. Findings Data from 93,622 participant-visits, including 4,702 deep-sequencing measurements, showed that the prevalence of NNRTI resistance among pre-treatment viremic PLHIV doubled between 2012 and 2017 (PR:1.98, 95%CI:1.34-2.91), rising to 9.61% (7.27-12.7%). The overall population prevalence of pre-treatment viremic NNRTI and NRTI resistance among all participants decreased during the same period, reaching 0.25% (0.18% - 0.33%) and 0.05% (0.02% - 0.10%), respectively (p-values for trend = 0.00015, 0.002), coincident with increasing treatment coverage and viral suppression. By the final survey, population prevalence of resistance contributed by treatment-experienced PLHIV exceeded that from pre-treatment PLHIV, with NNRTI resistance at 0.54% (0.44%-0.66%) and NRTI resistance at 0.42% (0.33%-0.53%). Overall, NNRTI and NRTI resistance was predominantly attributable to rtK103N and rtM184V. While 10.52% (7.97%-13.87%) and 9.95% (6.41%-15.43%) of viremic pre-treatment and treatment-experienced PLHIV harbored the inT97A mutation, no major dolutegravir resistance mutations were observed. Interpretation Despite rising NNRTI resistance among pre-treatment PLHIV, overall population prevalence of pre-treatment resistance decreased due to treatment uptake. Most NNRTI and NRTI resistance is now contributed by treatment-experienced PLHIV. The high prevalence of mutations conferring resistance to components of current first-line ART regimens among PLHIV with viremia is potentially concerning.
Collapse
Affiliation(s)
- Michael A. Martin
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Steven James Reynolds
- Rakai Health Sciences Program, Kalisizo, Uganda
- Division of Infectious Disease, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brian T. Foley
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Thomas C. Quinn
- Rakai Health Sciences Program, Kalisizo, Uganda
- Division of Infectious Disease, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven A. Kemp
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | | | - Ravindra K. Gupta
- Department of Medicine, University of Cambridge, Cambridge, UK
- Africa Health Research Institute, KwaZulu-Natal, South Africa
| | - Lucie Abeler-Dörner
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Joseph Kagaayi
- Rakai Health Sciences Program, Kalisizo, Uganda
- Makerere University School of Public Health, Kampala, Uganda
| | - Oliver Ratmann
- Department of Mathematics, Imperial College London, London, England, United Kingdom
| | - Christophe Fraser
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - David Bonsall
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - M. Kate Grabowski
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Rakai Health Sciences Program, Kalisizo, Uganda
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
16
|
Reddy N, Papathanasopoulos M, Steegen K, Basson AE. K103N, V106M and Y188L Significantly Reduce HIV-1 Subtype C Phenotypic Susceptibility to Doravirine. Viruses 2024; 16:1493. [PMID: 39339969 PMCID: PMC11437401 DOI: 10.3390/v16091493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Doravirine (DOR) is a non-nucleoside reverse transcriptase inhibitor (NNRTI) with efficacy against some NNRTI-resistant mutants. Although DOR resistance mutations are established for HIV-1 subtype B, it is less clear for non-B subtypes. This study investigated prevalent NNRTI resistance mutations on DOR susceptibility in HIV-1 subtype C. Prevalent drug resistance mutations were identified from a South African genotypic drug resistance testing database. Mutations, single or in combination, were introduced into replication-defective pseudoviruses and assessed for DOR susceptibility in vitro. The single V106M and Y188L mutations caused high-level resistance while others did not significantly impact DOR susceptibility. We observed an agreement between our in vitro and the Stanford HIVdb predicted susceptibilities. However, the F227L mutation was predicted to cause high-level DOR resistance but was susceptible in vitro. Combinations of mutations containing K103N, V106M or Y188L caused high-level resistance, in agreement with the predictions. These mutations are frequently observed in patients failing efavirenz- or nevirapine-based first-line regimens. However, they are also observed in those failing a protease inhibitor-based second-line regimen, as we have observed in our database. Genotypic drug resistance testing is therefore vital prior to the initiation of DOR-based treatment for those previously exposed to efavirenz or nevirapine.
Collapse
Affiliation(s)
- Nikita Reddy
- HIV Pathogenesis Research Unit, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Maria Papathanasopoulos
- HIV Pathogenesis Research Unit, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Kim Steegen
- Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Science, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Adriaan Erasmus Basson
- HIV Pathogenesis Research Unit, University of the Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
17
|
Charles OJ, Venturini C, Goldstein RA, Breuer J. HerpesDRG: a comprehensive resource for human herpesvirus antiviral drug resistance genotyping. BMC Bioinformatics 2024; 25:279. [PMID: 39192205 DOI: 10.1186/s12859-024-05885-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
The prevention and treatment of many herpesvirus associated diseases is based on the utilization of antiviral therapies, however therapeutic success is limited by the development of drug resistance. Currently no single database cataloguing resistance mutations exists, which hampers the use of sequence data for patient management. We therefore developed HerpesDRG, a drug resistance mutation database that incorporates all the known resistance genes and current treatment options, built from a systematic review of available genotype to phenotype literature. The database is released along with an R package that provides a simple approach to resistance variant annotation and clinical implication analysis from common sanger and next generation sequencing data. This represents the first openly available and community maintainable database of drug resistance mutations for the human herpesviruses (HHV), developed for the community of researchers and clinicians tackling HHV drug resistance.
Collapse
Affiliation(s)
- O J Charles
- Department of Infection, Immunity and Inflammation, University College London, Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK.
| | - C Venturini
- Department of Infection, Immunity and Inflammation, University College London, Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - R A Goldstein
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK
| | - J Breuer
- Department of Infection, Immunity and Inflammation, University College London, Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
- Great Ormond Street Hospital for Children National Health Service Foundation Trust, London, WC1N 1LE, UK
| |
Collapse
|
18
|
Beck IA, Boyce CL, Bishop MD, Vu YL, Fung A, Styrchak S, Panpradist N, Lutz BR, Frenkel LM. Development and Optimization of Oligonucleotide Ligation Assay (OLA) Probes for Detection of HIV-1 Resistance to Dolutegravir. Viruses 2024; 16:1162. [PMID: 39066324 PMCID: PMC11281587 DOI: 10.3390/v16071162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The WHO currently recommends dolutegravir (DTG)-based ART for persons living with HIV infection in resource-limited-settings (RLS). To expand access to testing for HIV drug resistance (DR) to DTG in RLS, we developed probes for use in the oligonucleotide ligation assay (OLA)-Simple, a near-point of care HIV DR kit. Genotypic data from clinical trials and case reports were used to determine the mutations in HIV-1 integrase critical to identifying individuals with DTG-resistance at virologic failure of DTG-based ART. Probes to detect G118R, Q148H/K/R, N155H and R263K in HIV-1 subtypes A, B, C, D and CRF01_AE were designed using sequence alignments from the Los Alamos database and validated using 61 clinical samples of HIV-1 subtypes A, B, C, D, CRF01_AE genotyped by PacBio (n = 15) or Sanger (n = 46). Initial OLA probes failed to ligate for 16/244 (6.5%) codons (9 at G118R and 7 at Q148H/K/R). Probes revised to accommodate polymorphisms interfering with ligation at codons G118R and Q148R reduced indeterminates to 3.7% (5 at G118R and 4 at Q148H/K/R) and detected DTG-mutations with a sensitivity of 96.5% and 100% specificity. These OLA DTG resistance probes appear highly sensitive and specific across HIV-1 subtypes common in RLS with high burden of HIV infection.
Collapse
Affiliation(s)
- Ingrid A. Beck
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA; (I.A.B.); (C.L.B.); (M.D.B.); (S.S.)
| | - Ceejay L. Boyce
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA; (I.A.B.); (C.L.B.); (M.D.B.); (S.S.)
| | - Marley D. Bishop
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA; (I.A.B.); (C.L.B.); (M.D.B.); (S.S.)
| | - Yen L. Vu
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; (Y.L.V.); (A.F.); (N.P.); (B.R.L.)
| | - Amanda Fung
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; (Y.L.V.); (A.F.); (N.P.); (B.R.L.)
| | - Sheila Styrchak
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA; (I.A.B.); (C.L.B.); (M.D.B.); (S.S.)
| | - Nuttada Panpradist
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; (Y.L.V.); (A.F.); (N.P.); (B.R.L.)
| | - Barry R. Lutz
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; (Y.L.V.); (A.F.); (N.P.); (B.R.L.)
| | - Lisa M. Frenkel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98109, USA; (I.A.B.); (C.L.B.); (M.D.B.); (S.S.)
- Departments of Medicine, Pediatrics and Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
19
|
Kanyerezi S, Sserwadda I, Ssemaganda A, Seruyange J, Ayitewala A, Oundo HR, Tenywa W, Kagurusi BA, Tusabe G, Were S, Ssewanyana I, Nabadda S, Namaganda MM, Mboowa G. HIV-DRIVES: HIV drug resistance identification, variant evaluation, and surveillance pipeline. Access Microbiol 2024; 6:000815.v3. [PMID: 39130735 PMCID: PMC11316571 DOI: 10.1099/acmi.0.000815.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/26/2024] [Indexed: 08/13/2024] Open
Abstract
The global prevalence of resistance to antiviral drugs combined with antiretroviral therapy (cART) emphasizes the need for continuous monitoring to better understand the dynamics of drug-resistant mutations to guide treatment optimization and patient management as well as check the spread of resistant viral strains. We have recently integrated next-generation sequencing (NGS) into routine HIV drug resistance (HIVDR) monitoring, with key challenges in the bioinformatic analysis and interpretation of the complex data generated, while ensuring data security and privacy for patient information. To address these challenges, here we present HIV-DRIVES (HIV Drug Resistance Identification, Variant Evaluation, and Surveillance), an NGS-HIVDR bioinformatics pipeline that has been developed and validated using Illumina short reads, FASTA, and Sanger ab1.seq files.
Collapse
Affiliation(s)
- Stephen Kanyerezi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O Box 7072 Kampala, Uganda
- The African Center of Excellence in Bioinformatics and Data-Intensive Sciences, the Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418 Kampala, Uganda
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Ivan Sserwadda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O Box 7072 Kampala, Uganda
- The African Center of Excellence in Bioinformatics and Data-Intensive Sciences, the Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418 Kampala, Uganda
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
- Amsterdam Institute for Global Health and Development (AIGHD), Department of Global Health, Academic Medical Center, Amsterdam, Netherlands
| | - Aloysious Ssemaganda
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Julius Seruyange
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Alisen Ayitewala
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Hellen Rosette Oundo
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Wilson Tenywa
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Brian A. Kagurusi
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Godwin Tusabe
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Stacy Were
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Isaac Ssewanyana
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Susan Nabadda
- National Health Laboratories and Diagnostics Services, Central Public Health Laboratories, Ministry of Health, P.O Box 7272 Kampala, Uganda
| | - Maria Magdalene Namaganda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O Box 7072 Kampala, Uganda
- The African Center of Excellence in Bioinformatics and Data-Intensive Sciences, the Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418 Kampala, Uganda
| | - Gerald Mboowa
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O Box 7072 Kampala, Uganda
- The African Center of Excellence in Bioinformatics and Data-Intensive Sciences, the Infectious Diseases Institute, College of Health Sciences, Makerere University, P.O Box 22418 Kampala, Uganda
- Africa Centres for Disease Control and Prevention, African Union Commission, Roosevelt Street, P.O. Box 3243, W21 K19 Addis Ababa, Ethiopia
| |
Collapse
|
20
|
Matthews LT, Jaggernath M, Kriel Y, Smith PM, Haberer JE, Baeten JM, Hendrix CW, Ware NC, Moodley P, Pillay M, Bennett K, Bassler J, Psaros C, Hurwitz KE, Bangsberg DR, Smit JA. Oral preexposure prophylaxis uptake, adherence, and persistence during periconception periods among women in South Africa. AIDS 2024; 38:1342-1354. [PMID: 38752557 PMCID: PMC11211057 DOI: 10.1097/qad.0000000000003925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 05/28/2024]
Abstract
OBJECTIVE We developed the Healthy Families-PrEP intervention to support HIV-prevention during periconception and pregnancy. We evaluated preexposure prophylaxis (PrEP) use with three objective measures. DESIGN This single-arm intervention study enrolled women in KwaZulu-Natal, South Africa, who were HIV-uninfected, not pregnant, in a relationship with a partner with HIV or unknown-serostatus, and with pregnancy plans. PrEP was offered as part of a comprehensive HIV prevention intervention. Participants were followed for 12 months. METHODS We evaluated periconception PrEP uptake and adherence using quarterly plasma tenofovir concentrations. We modeled factors associated with PrEP uptake and high plasma tenofovir (past day dosing). Patterns of use were analyzed using electronic pillcap data. Dried blood spots to measure intracellular tenofovir product (past 2 months dosing) were analyzed for a subset of women. RESULTS Three hundred thirty women with median age 24 (IQR: 22-27) years enrolled. Partner HIV-serostatus was unknown by 96% ( N = 316); 60% (195) initiated PrEP. High plasma tenofovir concentrations were seen in 35, 25, 22, and 20% of samples at 3, 6, 9, and 12 months, respectively. Similar adherence was measured by pillcap and dried blood spots. In adjusted models, lower income, alcohol use, and higher HIV stigma were associated with high plasma tenofovir. Eleven HIV-seroconversions were observed (incidence rate: 4.04/100 person-years [95% confidence interval: 2.24-7.30]). None had detectable plasma tenofovir. CONCLUSION The Healthy Families-PrEP intervention supported women in PrEP use. We observed high interest in periconception PrEP and over one-third adhered to PrEP in the first quarter; one-fifth were adherent over a year. High HIV incidence highlights the importance of strategies to reduce HIV incidence among periconception women. CLINICAL TRIAL NUMBER NCT03194308.
Collapse
Affiliation(s)
- Lynn T. Matthews
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manjeetha Jaggernath
- Maternal Adolescent and Child Health Research Unit (MRU), Department of Obstetrics and Gynaecology, University of the Witwatersrand, Faculty of Health Sciences, Durban, South Africa
| | - Yolandie Kriel
- Maternal Adolescent and Child Health Research Unit (MRU), Department of Obstetrics and Gynaecology, University of the Witwatersrand, Faculty of Health Sciences, Durban, South Africa
| | - Patricia M. Smith
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jessica E. Haberer
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jared M. Baeten
- Department of Global Health
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington
| | - Craig W. Hendrix
- Department of Medicine (Clinical Pharmacology), Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Norma C. Ware
- Department of Global Health & Social Medicine, Harvard Medical School
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Pravi Moodley
- University of KwaZulu-Natal, School of Laboratory Medicine and Medical Sciences, National Health Laboratory Service
| | - Melendhran Pillay
- Department of Virology, National Health Laboratory Service, Durban, South Africa
| | | | - John Bassler
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Christina Psaros
- Department of Psychiatry, Behavioural Medicine Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | - Jennifer A. Smit
- Maternal Adolescent and Child Health Research Unit (MRU), Department of Obstetrics and Gynaecology, University of the Witwatersrand, Faculty of Health Sciences, Durban, South Africa
| |
Collapse
|
21
|
Rashid A, Kang L, Yi F, Chu Q, Shah SA, Mahmood SF, Getaneh Y, Wei M, Chang S, Abidi SH, Shao Y. Human Immunodeficiency Virus Type-1 Genetic Diversity and Drugs Resistance Mutations among People Living with HIV in Karachi, Pakistan. Viruses 2024; 16:962. [PMID: 38932254 PMCID: PMC11209141 DOI: 10.3390/v16060962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
The human immunodeficiency virus type-1 epidemic in Pakistan has significantly increased over the last two decades. In Karachi, Pakistan, there is a lack of updated information on the complexity of HIV-1 genetic diversity and the burden of drug resistance mutations (DRMs) that can contribute to ART failure and poor treatment outcomes. This study aimed to determine HIV-1 genetic diversity and identify drug-resistance mutations among people living with HIV in Karachi. A total of 364 HIV-positive individuals, with a median age of 36 years, were enrolled in the study. The HIV-1 partial pol gene was successfully sequenced from 268 individuals. The sequences were used to generate phylogenetic trees to determine clade diversity and also to assess the burden of DRMs. Based on the partial pol sequences, 13 distinct HIV-1 subtypes and recombinant forms were identified. Subtype A1 was the most common clade (40%), followed by CRF02_AG (33.2%). Acquired DRMs were found in 30.6% of the ART-experienced patients, of whom 70.7%, 20.7%, and 8.5% were associated with resistance to NNRTIs, NRTIs, and PIs, respectively. Transmitted DRMs were found in 5.6% of the ART-naïve patients, of whom 93% were associated with resistance against NNRTIs and 7% to PIs. The high prevalence of DRMs in ART-experienced patients poses significant challenges to the long-term benefits and sustainability of the ART program. This study emphasizes the importance of continuous HIV molecular epidemiology and drug resistance surveillance to support evidence-based HIV prevention, precise ART, and targeted AIDS care.
Collapse
Affiliation(s)
- Abdur Rashid
- School of Medicine, Nankai University, Tianjin 300071, China; (A.R.); (M.W.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.K.); (Q.C.); (S.C.)
| | - Li Kang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.K.); (Q.C.); (S.C.)
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Feng Yi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.K.); (Q.C.); (S.C.)
| | - Qingfei Chu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.K.); (Q.C.); (S.C.)
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China;
| | | | | | - Yimam Getaneh
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China;
- Ethiopian Public Health Institute, Addis Ababa P.O. Box 1242, Ethiopia
| | - Min Wei
- School of Medicine, Nankai University, Tianjin 300071, China; (A.R.); (M.W.)
| | - Song Chang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.K.); (Q.C.); (S.C.)
| | - Syed Hani Abidi
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | - Yiming Shao
- School of Medicine, Nankai University, Tianjin 300071, China; (A.R.); (M.W.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (L.K.); (Q.C.); (S.C.)
- College of Life Sciences, Nankai University, Tianjin 300071, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China;
- Changping Laboratory, Yard 28, Science Park Road, Changping District, Beijing 102206, China
| |
Collapse
|
22
|
Wattanasombat S, Tongjai S. Easing genomic surveillance: A comprehensive performance evaluation of long-read assemblers across multi-strain mixture data of HIV-1 and Other pathogenic viruses for constructing a user-friendly bioinformatic pipeline. F1000Res 2024; 13:556. [PMID: 38984017 PMCID: PMC11231628 DOI: 10.12688/f1000research.149577.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 07/11/2024] Open
Abstract
Background Determining the appropriate computational requirements and software performance is essential for efficient genomic surveillance. The lack of standardized benchmarking complicates software selection, especially with limited resources. Methods We developed a containerized benchmarking pipeline to evaluate seven long-read assemblers-Canu, GoldRush, MetaFlye, Strainline, HaploDMF, iGDA, and RVHaplo-for viral haplotype reconstruction, using both simulated and experimental Oxford Nanopore sequencing data of HIV-1 and other viruses. Benchmarking was conducted on three computational systems to assess each assembler's performance, utilizing QUAST and BLASTN for quality assessment. Results Our findings show that assembler choice significantly impacts assembly time, with CPU and memory usage having minimal effect. Assembler selection also influences the size of the contigs, with a minimum read length of 2,000 nucleotides required for quality assembly. A 4,000-nucleotide read length improves quality further. Canu was efficient among de novo assemblers but not suitable for multi-strain mixtures, while GoldRush produced only consensus assemblies. Strainline and MetaFlye were suitable for metagenomic sequencing data, with Strainline requiring high memory and MetaFlye operable on low-specification machines. Among reference-based assemblers, iGDA had high error rates, RVHaplo showed the best runtime and accuracy but became ineffective with similar sequences, and HaploDMF, utilizing machine learning, had fewer errors with a slightly longer runtime. Conclusions The HIV-64148 pipeline, containerized using Docker, facilitates easy deployment and offers flexibility to select from a range of assemblers to match computational systems or study requirements. This tool aids in genome assembly and provides valuable information on HIV-1 sequences, enhancing viral evolution monitoring and understanding.
Collapse
Affiliation(s)
- Sara Wattanasombat
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siripong Tongjai
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
23
|
Tabler CO, Wegman SJ, Alhusaini N, Lee NF, Tilton JC. Premature Activation of the HIV-1 Protease Is Influenced by Polymorphisms in the Hinge Region. Viruses 2024; 16:849. [PMID: 38932142 PMCID: PMC11209583 DOI: 10.3390/v16060849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
HIV-1 protease inhibitors are an essential component of antiretroviral therapy. However, drug resistance is a pervasive issue motivating a persistent search for novel therapies. Recent reports found that when protease activates within the host cell's cytosol, it facilitates the pyroptotic killing of infected cells. This has led to speculation that promoting protease activation, rather than inhibiting it, could help to eradicate infected cells and potentially cure HIV-1 infection. Here, we used a nanoscale flow cytometry-based assay to characterize protease resistance mutations and polymorphisms. We quantified protease activity, viral concentration, and premature protease activation and confirmed previous findings that major resistance mutations generally destabilize the protease structure. Intriguingly, we found evidence that common polymorphisms in the hinge domain of protease can influence its susceptibility to premature activation. This suggests that viral heterogeneity could pose a considerable challenge for therapeutic strategies aimed at inducing premature protease activation in the future.
Collapse
Affiliation(s)
| | | | | | | | - John C. Tilton
- Center for Proteomics and Bioinformatics, Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (C.O.T.); (N.A.)
| |
Collapse
|
24
|
Chen GJ, Cheng CY, Yang CJ, Lee NY, Tang HJ, Huang SH, Huang MH, Liou BH, Lee YC, Lin CY, Hung TC, Lin SP, Sun HY, Chang SY, Hung CC. Trends of pre-treatment drug resistance in antiretroviral-naïve people with HIV-1 in the era of second-generation integrase strand-transfer inhibitors in Taiwan. J Antimicrob Chemother 2024; 79:1157-1163. [PMID: 38546761 DOI: 10.1093/jac/dkae086] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/06/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Monitoring the trends of pre-treatment drug resistance (PDR) and resistance-associated mutations (RAMs) among antiretroviral-naïve people with HIV (PWH) is important for the implementation of HIV treatment and control programmes. We analysed the trends of HIV-1 PDR after the introduction of second-generation integrase strand-transfer inhibitors (INSTIs) in 2016 in Taiwan, when single-tablet regimens of non-nucleoside reverse-transcriptase inhibitor (NNRTI-) and INSTI-based antiretroviral therapy became the preferred treatments. MATERIALS AND METHODS In this multicentre study, we included newly diagnosed, antiretroviral-naïve PWH who underwent tests for RAMs between 2016 and 2022. Pre-treatment genotypic resistance testing was performed, along with HIV-1 subtyping and determinations of plasma HIV RNA load and CD4 lymphocyte counts. RAMs were analysed using the Stanford University HIV Drug Resistance Database and only RAMs conferring at least low-level resistance were included. RESULTS From 2016 to 2022, pre-treatment blood samples from 3001 newly diagnosed PWH, which constituted 24.3% of newly diagnosed PWH in Taiwan during the study period, were tested. Of the PWH with analysable gene sequences, the HIV-1 PDR prevalence to NNRTIs, nucleoside reverse-transcriptase inhibitors (NRTIs), first- and second-generation INSTIs and PIs was 10.0%, 2.1%, 2.5%, 0.6% and 0.4%, respectively. While the trends of PDR remained stable for NRTIs, INSTIs and PIs, there was a significantly increasing trend of PDR to NNRTIs from 6.0% in 2016% to 13.1% in 2022 (P = 0.001). CONCLUSIONS After the introduction of second-generation INSTIs in Taiwan, the trends of HIV-1 PDR to NRTIs and INSTIs remained low. Furthermore, there was no significant decrease of the prevalence of PDR toward NNRTIs between 2016 and 2022.
Collapse
Affiliation(s)
- Guan-Jhou Chen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Infection Control Room, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Chien-Yu Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
- School of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Jui Yang
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Nan-Yao Lee
- Department of Internal Medicine, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Hung-Jen Tang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Sung-Hsi Huang
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
- Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Miao-Hui Huang
- Department of Internal Medicine, Hualien Tzu Chi Hospital and Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Bo-Huang Liou
- Department of Internal Medicine, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Yi-Chien Lee
- Department of Internal Medicine, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Chi-Ying Lin
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Tung-Che Hung
- Department of Infectious Diseases, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Shih-Ping Lin
- Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsin-Yun Sun
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chien-Ching Hung
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| |
Collapse
|
25
|
Cluck DB, Chastain DB, Murray M, Durham SH, Chahine EB, Derrick C, Dumond JB, Hester EK, Jeter SB, Johnson MD, Kilcrease C, Kufel WD, Kwong J, Ladak AF, Patel N, Pérez SE, Poe JB, Bolch C, Thomas I, Asiago-Reddy E, Short WR. Consensus recommendations for the use of novel antiretrovirals in persons with HIV who are heavily treatment-experienced and/or have multidrug-resistant HIV-1: Endorsed by the American Academy of HIV Medicine, American College of Clinical Pharmacy. Pharmacotherapy 2024; 44:360-382. [PMID: 38853601 DOI: 10.1002/phar.2914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 06/11/2024]
Abstract
Treatment options are currently limited for persons with HIV-1 (PWH) who are heavily treatment-experienced and/or have multidrug-resistant HIV-1. Three agents have been approved by the U.S. Food and Drug Administration (FDA) since 2018, representing a significant advancement for this population: ibalizumab, fostemsavir, and lenacapavir. However, there is a paucity of recommendations endorsed by national and international guidelines describing the optimal use (e.g., selection and monitoring after initiation) of these novel antiretrovirals in this population. To address this gap, a modified Delphi technique was used to develop these consensus recommendations that establish a framework for initiating and managing ibalizumab, fostemsavir, or lenacapavir in PWH who are heavily treatment-experienced and/or have multidrug-resistant HIV-1. In addition, future areas of research are also identified and discussed.
Collapse
Affiliation(s)
- David B Cluck
- Department of Pharmacy Practice, East Tennessee State University Bill Gatton College of Pharmacy, Johnson City, Tennessee, USA
| | | | - Milena Murray
- Midwestern University College of Pharmacy, Downers Grove, Illinois, USA
- Northwestern Medicine, Evanston, Illinois, USA
| | - Spencer H Durham
- Department of Pharmacy Practice, Auburn University Harrison College of Pharmacy, Auburn, Alabama, USA
| | - Elias B Chahine
- Department of Pharmacy Practice, Palm Beach Atlantic University Gregory School of Pharmacy, West Palm Beach, Florida, USA
| | | | - Julie B Dumond
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - E Kelly Hester
- Department of Pharmacy Practice, Auburn University Harrison College of Pharmacy, Auburn, Alabama, USA
| | - Sarah B Jeter
- University of Kentucky HealthCare, Lexington, Kentucky, USA
| | | | - Christin Kilcrease
- HIV Prevention/Treatment and Primary Care, The Johns Hopkins Hospital, John G. Bartlett Specialty Practice, Baltimore, Maryland, USA
| | - Wesley D Kufel
- Binghamton University School of Pharmacy and Pharmaceutical Sciences, Binghamton, New York, USA
- Division of Infectious Diseases, State University of New York Upstate Medical University, Syracuse, New York, USA
- State University of New York Upstate University Hospital, Syracuse, New York, USA
| | - Jeffrey Kwong
- Division of Advanced Practice, School of Nursing, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Amber F Ladak
- Ryan White Program, Division of Infectious Disease, Augusta University, Augusta, Georgia, USA
| | - Nimish Patel
- Division of Clinical Pharmacy, Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Sarah E Pérez
- HIV and Primary Care, Ruth M. Rothstein CORE Center, Chicago, Illinois, USA
| | - Jonell B Poe
- Ryan White Program, Division of Infectious Disease, Augusta University, Augusta, Georgia, USA
- School of Allied Health, Augusta University, Augusta, Georgia, USA
- Department of Psychiatry, HIV/LBTGQ Behavioral Track, Augusta University, Augusta, Georgia, USA
| | - Charlotte Bolch
- Office of Research and Sponsored Programs, Midwestern University, Glendale, Arizona, USA
| | - Ian Thomas
- University of Georgia, Athens, Georgia, USA
| | - Elizabeth Asiago-Reddy
- Division of Infectious Diseases, State University of New York Upstate Medical University, Syracuse, New York, USA
- Inclusive Health Services, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - William R Short
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Hernández Berthet AS, Aptekmann AA, Tejero J, Sánchez IE, Noguera ME, Roman EA. Associating protein sequence positions with the modulation of quantitative phenotypes. Arch Biochem Biophys 2024; 755:109979. [PMID: 38583654 DOI: 10.1016/j.abb.2024.109979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/11/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
Although protein sequences encode the information for folding and function, understanding their link is not an easy task. Unluckily, the prediction of how specific amino acids contribute to these features is still considerably impaired. Here, we developed a simple algorithm that finds positions in a protein sequence with potential to modulate the studied quantitative phenotypes. From a few hundred protein sequences, we perform multiple sequence alignments, obtain the per-position pairwise differences for both the sequence and the observed phenotypes, and calculate the correlation between these last two quantities. We tested our methodology with four cases: archaeal Adenylate Kinases and the organisms optimal growth temperatures, microbial rhodopsins and their maximal absorption wavelengths, mammalian myoglobins and their muscular concentration, and inhibition of HIV protease clinical isolates by two different molecules. We found from 3 to 10 positions tightly associated with those phenotypes, depending on the studied case. We showed that these correlations appear using individual positions but an improvement is achieved when the most correlated positions are jointly analyzed. Noteworthy, we performed phenotype predictions using a simple linear model that links per-position divergences and differences in the observed phenotypes. Predictions are comparable to the state-of-art methodologies which, in most of the cases, are far more complex. All of the calculations are obtained at a very low information cost since the only input needed is a multiple sequence alignment of protein sequences with their associated quantitative phenotypes. The diversity of the explored systems makes our work a valuable tool to find sequence determinants of biological activity modulation and to predict various functional features for uncharacterized members of a protein family.
Collapse
Affiliation(s)
- Ayelén S Hernández Berthet
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Intendente Güiraldes 2160 - Ciudad Universitaria, 1428EGA, C.A.B.A., Argentina.
| | - Ariel A Aptekmann
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Laboratorio de Fisiología de Proteínas, Buenos Aires, Argentina; Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, 08873, USA; Institute of Marine and Coastal Sciences, Rutgers University, New Brunswick, NJ, 08901, USA.
| | - Jesús Tejero
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15260, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Ignacio E Sánchez
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Laboratorio de Fisiología de Proteínas, Buenos Aires, Argentina.
| | - Martín E Noguera
- Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Dr. Alejandro Paladini, Junín 956, 1113AAD, C.A.B.A., Argentina; Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Saenz Peña 352, B1876BXD, Bernal, Argentina.
| | - Ernesto A Roman
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Intendente Güiraldes 2160 - Ciudad Universitaria, 1428EGA, C.A.B.A., Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Dr. Alejandro Paladini, Junín 956, 1113AAD, C.A.B.A., Argentina.
| |
Collapse
|
27
|
Biswas A, Choudhuri I, Arnold E, Lyumkis D, Haldane A, Levy RM. Kinetic coevolutionary models predict the temporal emergence of HIV-1 resistance mutations under drug selection pressure. Proc Natl Acad Sci U S A 2024; 121:e2316662121. [PMID: 38557187 PMCID: PMC11009627 DOI: 10.1073/pnas.2316662121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
Drug resistance in HIV type 1 (HIV-1) is a pervasive problem that affects the lives of millions of people worldwide. Although records of drug-resistant mutations (DRMs) have been extensively tabulated within public repositories, our understanding of the evolutionary kinetics of DRMs and how they evolve together remains limited. Epistasis, the interaction between a DRM and other residues in HIV-1 protein sequences, is key to the temporal evolution of drug resistance. We use a Potts sequence-covariation statistical-energy model of HIV-1 protein fitness under drug selection pressure, which captures epistatic interactions between all positions, combined with kinetic Monte-Carlo simulations of sequence evolutionary trajectories, to explore the acquisition of DRMs as they arise in an ensemble of drug-naive patient protein sequences. We follow the time course of 52 DRMs in the enzymes protease, RT, and integrase, the primary targets of antiretroviral therapy. The rates at which DRMs emerge are highly correlated with their observed acquisition rates reported in the literature when drug pressure is applied. This result highlights the central role of epistasis in determining the kinetics governing DRM emergence. Whereas rapidly acquired DRMs begin to accumulate as soon as drug pressure is applied, slowly acquired DRMs are contingent on accessory mutations that appear only after prolonged drug pressure. We provide a foundation for using computational methods to determine the temporal evolution of drug resistance using Potts statistical potentials, which can be used to gain mechanistic insights into drug resistance pathways in HIV-1 and other infectious agents.
Collapse
Affiliation(s)
- Avik Biswas
- Center for Biophysics and Computational Biology, College of Science and Technology, Temple University, Philadelphia, PA19122
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA92037
- Department of Physics, University of California San Diego, La Jolla, CA92093
| | - Indrani Choudhuri
- Center for Biophysics and Computational Biology, College of Science and Technology, Temple University, Philadelphia, PA19122
- Department of Chemistry, Temple University, Philadelphia, PA19122
| | - Eddy Arnold
- Department of Chemistry and Chemical Biology, Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ08854
| | - Dmitry Lyumkis
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA92037
- Graduate School of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA92093
| | - Allan Haldane
- Center for Biophysics and Computational Biology, College of Science and Technology, Temple University, Philadelphia, PA19122
- Department of Physics, Temple University, Philadelphia, PA19122
| | - Ronald M. Levy
- Center for Biophysics and Computational Biology, College of Science and Technology, Temple University, Philadelphia, PA19122
- Department of Chemistry, Temple University, Philadelphia, PA19122
| |
Collapse
|
28
|
Wang Z. Novel diarylpyrimidine subtypes as HIV-1 nonnucleoside reverse transcriptase inhibitors with improved resistance profile. J Med Virol 2024; 96:e29553. [PMID: 38516803 DOI: 10.1002/jmv.29553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Affiliation(s)
- Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
29
|
Hikichi Y, Grover JR, Schäfer A, Mothes W, Freed EO. Epistatic pathways can drive HIV-1 escape from integrase strand transfer inhibitors. SCIENCE ADVANCES 2024; 10:eadn0042. [PMID: 38427738 PMCID: PMC10906922 DOI: 10.1126/sciadv.adn0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/26/2024] [Indexed: 03/03/2024]
Abstract
People living with human immunodeficiency virus (HIV) receiving integrase strand transfer inhibitors (INSTIs) have been reported to experience virological failure in the absence of resistance mutations in integrase. To elucidate INSTI resistance mechanisms, we propagated HIV-1 in the presence of escalating concentrations of the INSTI dolutegravir. HIV-1 became resistant to dolutegravir by sequentially acquiring mutations in the envelope glycoprotein (Env) and the nucleocapsid protein. The selected Env mutations enhance the ability of the virus to spread via cell-cell transfer, thereby increasing the multiplicity of infection (MOI). While the selected Env mutations confer broad resistance to multiple classes of antiretrovirals, the fold resistance is ~2 logs higher for INSTIs than for other classes of drugs. We demonstrate that INSTIs are more readily overwhelmed by high MOI than other classes of antiretrovirals. Our findings advance the understanding of how HIV-1 can evolve resistance to antiretrovirals, including the potent INSTIs, in the absence of drug-target gene mutations.
Collapse
Affiliation(s)
- Yuta Hikichi
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jonathan R. Grover
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Alicia Schäfer
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Eric O. Freed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
30
|
Ferreiro D, Khalil R, Sousa SF, Arenas M. Substitution Models of Protein Evolution with Selection on Enzymatic Activity. Mol Biol Evol 2024; 41:msae026. [PMID: 38314876 PMCID: PMC10873502 DOI: 10.1093/molbev/msae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/07/2024] Open
Abstract
Substitution models of evolution are necessary for diverse evolutionary analyses including phylogenetic tree and ancestral sequence reconstructions. At the protein level, empirical substitution models are traditionally used due to their simplicity, but they ignore the variability of substitution patterns among protein sites. Next, in order to improve the realism of the modeling of protein evolution, a series of structurally constrained substitution models were presented, but still they usually ignore constraints on the protein activity. Here, we present a substitution model of protein evolution with selection on both protein structure and enzymatic activity, and that can be applied to phylogenetics. In particular, the model considers the binding affinity of the enzyme-substrate complex as well as structural constraints that include the flexibility of structural flaps, hydrogen bonds, amino acids backbone radius of gyration, and solvent-accessible surface area that are quantified through molecular dynamics simulations. We applied the model to the HIV-1 protease and evaluated it by phylogenetic likelihood in comparison with the best-fitting empirical substitution model and a structurally constrained substitution model that ignores the enzymatic activity. We found that accounting for selection on the protein activity improves the fitting of the modeled functional regions with the real observations, especially in data with high molecular identity, which recommends considering constraints on the protein activity in the development of substitution models of evolution.
Collapse
Affiliation(s)
- David Ferreiro
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Department of Biochemistry, Genetics and Immunology, Universidade de Vigo, 36310 Vigo, Spain
| | - Ruqaiya Khalil
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Department of Biochemistry, Genetics and Immunology, Universidade de Vigo, 36310 Vigo, Spain
| | - Sergio F Sousa
- UCIBIO/REQUIMTE, BioSIM, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Miguel Arenas
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Department of Biochemistry, Genetics and Immunology, Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
31
|
Elén S, Björkman P, Zazzi M, Böhm M, Bernal E, Sönnerborg A, Elvstam O. Low-level HIV viraemia during antiretroviral therapy: Longitudinal patterns and predictors of viral suppression. HIV Med 2024; 25:107-116. [PMID: 37721192 DOI: 10.1111/hiv.13541] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVES Our objective was to characterize longitudinal patterns of viraemia and factors associated with viral suppression in people with HIV and low-level viraemia (LLV) during antiretroviral therapy (ART). METHODS We included people with HIV in the EuResist Integrated Database with LLV following ART initiation after 2005. LLV was defined as two or more consecutive viral load (VL) measurements of 51-199 copies/mL 30-365 days apart after >12 months of ART. Viraemia patterns were analyzed over 24 months. Factors associated with viral suppression at 12 months after LLV episodes were identified using univariable and multivariable logistic regression. RESULTS Of 25 113 people with HIV, 2474 (9.9%) had LLV. Among 1387 participants with 24 months of follow-up after LLV, 406 (29%) had persistent suppression, 669 (48%) had transient viraemic episodes, 29 (2%) had persistent LLV, and 283 (20%) had virological failure. Following LLV episodes, the proportion with detectable viraemia declined (p for trend <0.001 and 0.034, in the first and second year, respectively). At 12 months, 68% had undetectable VL, which was associated with suppression before LLV (adjusted odds ratio [aOR] 1.7; 95% confidence interval [CI] 1.2-2.4) and ART modification after LLV (aOR 1.6; 95% CI 1.0-2.4). The following factors were negatively associated with undetectable VL at 12 months: higher VL during LLV (aOR 0.57 per log10 copies/mL; 95% CI 0.37-0.89), injecting drug use (aOR 0.67; 95% CI 0.47-0.96), and regimens with protease inhibitors (aOR 0.65; 95% CI 0.49-0.87) or combined anchor drugs (aOR 0.52; 95% CI 0.32-0.85). CONCLUSION Most people with LLV did not experience sustained viral suppression during 24-month follow-up, supporting the association between LLV and inferior treatment outcome.
Collapse
Affiliation(s)
- S Elén
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - P Björkman
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Infectious Diseases, Skåne University Hospital, Malmö, Sweden
| | - M Zazzi
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - M Böhm
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - E Bernal
- Infectious Disease Unit. Reina Sofia Hospital, Murcia University and IMIB, Murcia, Spain
| | - A Sönnerborg
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - O Elvstam
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Infectious Diseases, Växjö Central Hospital, Växjö, Sweden
| |
Collapse
|
32
|
Zhou S, Long N, Moeser M, Hill CS, Samoff E, Mobley V, Frost S, Bayer C, Kelly E, Greifinger A, Shone S, Glover W, Clark M, Eron J, Cohen M, Swanstrom R, Dennis AM. Use of Next-Generation Sequencing in a State-Wide Strategy of HIV-1 Surveillance: Impact of the SARS-COV-2 Pandemic on HIV-1 Diagnosis and Transmission. J Infect Dis 2023; 228:1758-1765. [PMID: 37283544 PMCID: PMC10733719 DOI: 10.1093/infdis/jiad211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/24/2023] [Accepted: 06/05/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic posed an unpreceded threat to the management of other pandemics such as human immunodeficiency virus-1 (HIV-1) in the United States. The full impact of the SARS-CoV-2 pandemic on the HIV-1 pandemic needs to be evaluated. METHODS All individuals with newly reported HIV-1 diagnoses from NC State Laboratory of Public Health were enrolled in this prospective observational study, 2018-2021. We used a sequencing-based recency assay to identify recent HIV-1 infections and to determine the days postinfection (DPI) for each person at the time of diagnosis. RESULTS Sequencing used diagnostic serum samples from 814 individuals with new HIV-1 diagnoses spanning this 4-year period. Characteristics of individuals diagnosed in 2020 differed from those in other years. People of color diagnosed in 2021 were on average 6 months delayed in their diagnosis compared to those diagnosed in 2020. There was a trend that genetic networks were more known for individuals diagnosed in 2021. We observed no major integrase resistance mutations over the course of the study. CONCLUSIONS SARS-CoV-2 pandemic may contribute to the spread of HIV-1. Public health resources need to focus on restoring HIV-1 testing and interrupting active, ongoing, transmission.
Collapse
Affiliation(s)
- Shuntai Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nathan Long
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matt Moeser
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Collin S Hill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Erika Samoff
- North Carolina Department of Health and Human Services, Raleigh, North Carolina, USA
| | - Victoria Mobley
- North Carolina Department of Health and Human Services, Raleigh, North Carolina, USA
| | - Simon Frost
- Microsoft Health Futures, Microsoft Corporation, Redmond, Washington, USA
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Cara Bayer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Elizabeth Kelly
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Annalea Greifinger
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Scott Shone
- North Carolina Department of Health and Human Services, Raleigh, North Carolina, USA
| | - William Glover
- North Carolina Department of Health and Human Services, Raleigh, North Carolina, USA
| | - Michael Clark
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joseph Eron
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Myron Cohen
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ronald Swanstrom
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ann M Dennis
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
33
|
Henegar C, Letang E, Wang R, Hicks C, Fox D, Jones B, de Ruiter A, Vannappagari V. A Comprehensive Literature Review of Treatment-Emergent Integrase Resistance with Dolutegravir-Based Regimens in Real-World Settings. Viruses 2023; 15:2426. [PMID: 38140667 PMCID: PMC10747437 DOI: 10.3390/v15122426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
After a decade of dolutegravir (DTG) use in various antiretroviral therapy combinations and in diverse populations globally, it is critical to identify HIV strains with reduced drug susceptibility and monitor emergent resistance in people living with HIV who experience virologic failure while on DTG-based regimens. We searched the PubMed, Embase, and Cochrane databases to identify studies that reported DTG resistance-associated mutations (RAMs) emerging under selection pressure. Our review showed that RAMs conferring resistance to DTG were rare in 2-drug and 3-drug regimens used in real-world cohorts, corroborating data from clinical trials. The potency of DTG in maintaining virologic suppression was demonstrated, even in cases of pre-existing resistance to companion drugs in the regimen. Estimates of DTG RAMs depended on the population and certain risk factors, including monotherapy, baseline resistance or lack of genotypic testing, treatment history and prior virologic failure, and suboptimal treatment adherence. The RAMs detected after virologic failure, often in heavily treatment-experienced individuals with prior exposure to integrase strand transfer inhibitors, were G118R, E138K, G140A/C/R/S, Q148H/K/R, N155H, and R263K. Overall, these data highlight the durable effectiveness and high barrier to resistance of DTG as part of combination antiretroviral therapy in a wide variety of settings.
Collapse
Affiliation(s)
- Cassidy Henegar
- ViiV Healthcare, 406 Blackwell Street, Suite 300, Durham, NC 27701, USA; (R.W.); (C.H.); (D.F.); (V.V.)
| | - Emilio Letang
- ViiV Healthcare, P.T.M., Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Ruolan Wang
- ViiV Healthcare, 406 Blackwell Street, Suite 300, Durham, NC 27701, USA; (R.W.); (C.H.); (D.F.); (V.V.)
| | - Charles Hicks
- ViiV Healthcare, 406 Blackwell Street, Suite 300, Durham, NC 27701, USA; (R.W.); (C.H.); (D.F.); (V.V.)
| | - Dainielle Fox
- ViiV Healthcare, 406 Blackwell Street, Suite 300, Durham, NC 27701, USA; (R.W.); (C.H.); (D.F.); (V.V.)
| | - Bryn Jones
- ViiV Healthcare, 980 Great West Road, Brentford TW8 9GS, Middlesex, UK
| | | | - Vani Vannappagari
- ViiV Healthcare, 406 Blackwell Street, Suite 300, Durham, NC 27701, USA; (R.W.); (C.H.); (D.F.); (V.V.)
| |
Collapse
|
34
|
Jenkins F, Le T, Farhat R, Pinto A, Anzari A, Bonsall D, Golubchik T, Bowden R, Lee FJ, van Hal SJ. Validation of an HIV whole genome sequencing method for HIV drug resistance testing in an Australian clinical microbiology laboratory. J Med Virol 2023; 95:e29273. [PMID: 38050831 DOI: 10.1002/jmv.29273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 12/07/2023]
Abstract
Detection of HIV drug resistance (HIVDR) is vital to successful anti-retroviral therapy (ART). HIVDR testing to determine drug-resistance mutations is routinely performed in Australia to guide ART choice in newly diagnosed people living with HIV or in cases of treatment failure. In 2022, our clinical microbiology laboratory sought to validate a next-generation sequencing (NGS)-based HIVDR assay to replace the previous Sanger-sequencing (SS)-based ViroSeq. NGS solutions for HIVDR offer higher throughput, lower costs and higher sensitivity for variant detection. We sought to validate the previously described low-cost probe-based NGS method (veSEQ-HIV) for whole-genome recovery and HIVDR-testing in a diagnostic setting. veSEQ-HIV displayed 100% and 98% accuracy in major and minor mutation detection, respectively, and 100% accuracy of subtyping (provided > 1000 mapped reads were obtained). Pairwise comparison exhibited low inter-and intrarun variability across the whole-genome (Jaccard index [J] = 0.993; J = 0.972) and the Pol gene (J = 0.999; J = 0.999), respectively. veSEQ-HIV met all our pre-set criteria based on WHO recommendations and successfully replaced ViroSeq in our laboratory. Scaling-down veSEQ-HIV to a limited batch size and sequencing on Illumina iSeq. 100, allowed easy implementation of the assay into the workflow of a small sequencing laboratory with minimal staff and equipment and the ability to meet clinically relevant test turn-around times. As HIVDR-testing moves from SS- to NGS-based methods and new ART drugs come to market (particularly those with targets outside the Pol region), whole-genome recovery using veSEQ-HIV provides a robust, cost-effective and "future-proof" NGS method for HIVDR-testing.
Collapse
Affiliation(s)
- Frances Jenkins
- Department of Infectious Diseases and Microbiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Thomas Le
- Department of Infectious Diseases and Microbiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Rima Farhat
- Department of Infectious Diseases and Microbiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Angie Pinto
- Department of Infectious Diseases and Microbiology, Royal Prince Alfred Hospital, Sydney, Australia
- The Kirby Institute, UNSW Australia, Sydney, Australia
| | - Azim Anzari
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - David Bonsall
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Tanya Golubchik
- Department of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Rory Bowden
- The Walter and Eliza Hall Institute of Medical Research, Advanced Genomics Facility, Melbourne, Australia
| | - Frederick J Lee
- Department of Clinical Immunology and Allergy, Royal Prince Alfred Hospital, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Sebastiaan J van Hal
- Department of Infectious Diseases and Microbiology, Royal Prince Alfred Hospital, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
35
|
Antonova A, Kazennova E, Lebedev A, Ozhmegova E, Kuznetsova A, Tumanov A, Bobkova M. Recombinant Forms of HIV-1 in the Last Decade of the Epidemic in the Russian Federation. Viruses 2023; 15:2312. [PMID: 38140553 PMCID: PMC10748268 DOI: 10.3390/v15122312] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Currently, HIV-1 displays a substantial level of genetic diversity on a global scale, partly attributed to its recombinant variants. This study seeks to identify and analyze HIV-1 recombinants in Russia during the last decade of the epidemic. A comprehensive examination was conducted, encompassing 3178 partial pol sequences. Subtyping was achieved through various programs including COMET, the Stanford Database, REGA, jpHMM, RIP, and RDP4 for recombination analysis. The study also involved phylogenetic analysis to trace the origins of the identified recombinants. Primary resistance (PrimDR) prevalence and Drug Resistance Mutations (DRMs) were assessed. The study uncovered an overall proportion of recombinants at 8.7%, with a statistically significant increase in their frequency observed over time (p < 0.001). The Northwestern (18.5%) and Siberian (15.0%) Federal Districts exhibited a high prevalence of recombinants, while the Volga (1.9%) and Ural (2.8%) Federal Districts had a lower prevalence. Among HIV-1 recombinants, a PrimDR prevalence of 11.4% was identified. Notably, significant differences in DRMs were observed, with a higher prevalence of M184V in sub-subtype A6 (p = 0.018) and K103N in CRF63_02A6 (p = 0.002). These findings underscore the increasing HIV-1 genetic diversity and highlight a substantial prevalence of PrimDR among its recombinant forms, emphasizing the necessity for ongoing systematic monitoring.
Collapse
Affiliation(s)
- Anastasiia Antonova
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.K.); (A.L.); (E.O.); (A.K.); (A.T.)
| | - Elena Kazennova
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.K.); (A.L.); (E.O.); (A.K.); (A.T.)
| | - Aleksey Lebedev
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.K.); (A.L.); (E.O.); (A.K.); (A.T.)
| | - Ekaterina Ozhmegova
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.K.); (A.L.); (E.O.); (A.K.); (A.T.)
| | - Anna Kuznetsova
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.K.); (A.L.); (E.O.); (A.K.); (A.T.)
| | - Aleksandr Tumanov
- The National Research Center for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (E.K.); (A.L.); (E.O.); (A.K.); (A.T.)
| | - Marina Bobkova
- I. Mechnikov Research Institute for Vaccines and Sera, 105064 Moscow, Russia;
| |
Collapse
|
36
|
Krishnan S, Lippincott CK, Bjerrum S, Martinez Rivera MB, Shah M. A National HIV Provider Survey of Antiretroviral Therapy Preferences for Management of Treatment-Naive and Experienced Individuals With Drug Resistance. Open Forum Infect Dis 2023; 10:ofad541. [PMID: 38023561 PMCID: PMC10655941 DOI: 10.1093/ofid/ofad541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Background HIV clinical practice guidelines outline broad treatment principles but offer less explicit recommendations by permutations of encountered viral resistance. We hypothesize that there is variability in antiretroviral (ARV) regimen decision making among providers when considering HIV drug resistance (HIVDR). Methods US HIV providers provided ARV regimen recommendations for case vignettes in a series of electronic surveys encompassing variations of HIVDR. Responses were characterized by drugs and classes selected and anticipated activity based on genotypic susceptibility. Heterogeneity was defined as the proportion of unique ARV regimens from total responses. Results An overall 119 providers from the United States participated. Among case vignettes with isolated M184V and viremia, 85.9% selected a regimen with 2 nucleoside reverse transcriptase inhibitors (NRTIs) + integrase strand transfer inhibitor (INSTI); 9.9% selected regimens with >3 ARVs. Alternatively, in scenarios of viremia with moderate to high-level NRTI resistance, >50% of providers selected an NRTI-sparing regimen, while a minority recommended 2 NRTIs + INSTI (21/123, 17%). In moderate to high-level INSTI resistance, there was response heterogeneity, with no common unifying approach to management (127 unique regimens/181 responses, 70% heterogeneity). Providers used cabotegravir/rilpivirine for treatment simplification in suppressed cases, despite a history of treatment failure (37/205, 36%). Conclusions Our national survey of US HIV providers revealed a consensus to management of HIV resistance with potential alternative options in cases with low heterogeneity. Providers selected cabotegravir/rilpivirine as a viable treatment simplification strategy in suppressed cases with a history of treatment failure. The responses to the case vignettes could be used an education tool for ARV decision making in HIVDR.
Collapse
Affiliation(s)
- Sonya Krishnan
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher K Lippincott
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Stephanie Bjerrum
- Department of Infectious Diseases, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Marina B Martinez Rivera
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Maunank Shah
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
37
|
Ditz JC, Reuter B, Pfeifer N. Inherently interpretable position-aware convolutional motif kernel networks for biological sequencing data. Sci Rep 2023; 13:17216. [PMID: 37821530 PMCID: PMC10567796 DOI: 10.1038/s41598-023-44175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023] Open
Abstract
Artificial neural networks show promising performance in detecting correlations within data that are associated with specific outcomes. However, the black-box nature of such models can hinder the knowledge advancement in research fields by obscuring the decision process and preventing scientist to fully conceptualize predicted outcomes. Furthermore, domain experts like healthcare providers need explainable predictions to assess whether a predicted outcome can be trusted in high stakes scenarios and to help them integrating a model into their own routine. Therefore, interpretable models play a crucial role for the incorporation of machine learning into high stakes scenarios like healthcare. In this paper we introduce Convolutional Motif Kernel Networks, a neural network architecture that involves learning a feature representation within a subspace of the reproducing kernel Hilbert space of the position-aware motif kernel function. The resulting model enables to directly interpret and evaluate prediction outcomes by providing a biologically and medically meaningful explanation without the need for additional post-hoc analysis. We show that our model is able to robustly learn on small datasets and reaches state-of-the-art performance on relevant healthcare prediction tasks. Our proposed method can be utilized on DNA and protein sequences. Furthermore, we show that the proposed method learns biologically meaningful concepts directly from data using an end-to-end learning scheme.
Collapse
Affiliation(s)
- Jonas C Ditz
- Methods in Medical Informatics, Department of Computer Science, University of Tübingen, Sand 14, Tübingen, 72076, Germany.
| | - Bernhard Reuter
- Methods in Medical Informatics, Department of Computer Science, University of Tübingen, Sand 14, Tübingen, 72076, Germany
| | - Nico Pfeifer
- Methods in Medical Informatics, Department of Computer Science, University of Tübingen, Sand 14, Tübingen, 72076, Germany.
| |
Collapse
|
38
|
Chen GJ, Sun HY, Chang SY, Hsieh SM, Sheng WH, Chuang YC, Huang YS, Lin KY, Liu WC, Su YC, Hung CC. Effectiveness of second-generation integrase strand-transfer inhibitor-based regimens for antiretroviral-experienced people with HIV who had viral rebound. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:988-995. [PMID: 37574435 DOI: 10.1016/j.jmii.2023.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/22/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Antiretroviral regimens containing a second-generation integrase strand-transfer inhibitor (INSTI) plus 2 nucleos(t)ide reverse-transcriptase inhibitors (NRTIs) are the recommended therapy for people with HIV (PWH) who are antiretroviral-naïve or on stable antiretroviral therapy (ART) with viral suppression. Real-world data on the virologic effectiveness of co-formulated bictegravir/emtricitabine/tenofovir alafenamide (BIC/FTC/TAF) among PWH with virologic failure while receiving other ART remain sparse. METHODS We retrospectively reviewed the medical records of PWH who had viral rebound with plasma HIV RNA >1000 copies/mL and were switched to either dolutegravir combined with 2 NRTIs or BIC/FTC/TAF. The primary end point was re-achieving viral suppression within the first 48 weeks of switch. The association between NRTI-related resistance-associated mutations (RAMs) and virologic effectiveness was examined. RESULTS Seventy-nine PWH with viral rebound while receiving other antiretroviral regimens were included. Within the first 48 weeks of switch, the overall probability of re-achieving viral suppression was 79.7% (82.5% [33/40] and 76.9% [30/39] for BIC/FTC/TAF and dolutegravir-based regimens, respectively, p = 0.78). PWH with a higher CD4 lymphocyte count (adjusted odds ratio, per 100-cell/mm3 increase, 1.41; 95% confidence interval, 1.02-1.95) were more likely to re-achieve viral suppression. Among PWH switching to BIC/FTC/TAF who had pre-existing RAMs to NRTIs before switch, 14 of 15 (93.3%) successfully achieved viral suppression. CONCLUSIONS Switching to BIC/FTC/TAF and dolutegravir-based regimens could re-achieve viral suppression in four-fifth of the PWH who experienced viral rebound during treatment with other antiretroviral regimens. Pre-existing NRTI-related RAMs did not have adverse impact on the effectiveness of dolutegravir combined with 2 NRTIs or BIC/FTC/TAF.
Collapse
Affiliation(s)
- Guan-Jhou Chen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Hsin-Yun Sun
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sui-Yuan Chang
- Department of Laboratory Medicine, National Taiwan University Hospital and National Taiwan University College Medicine, Taipei, Taiwan; Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Szu-Min Hsieh
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wang-Hui Sheng
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Chung Chuang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Shan Huang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Yin Lin
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Chun Liu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Ching Su
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chien-Ching Hung
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan.
| |
Collapse
|
39
|
Cilento ME, Wen X, Reeve AB, Ukah OB, Snyder AA, Carrillo CM, Smith CP, Edwards K, Wahoski CC, Kitzler DR, Kodama EN, Mitsuya H, Parniak MA, Tedbury PR, Sarafianos SG. HIV-1 Resistance to Islatravir/Tenofovir Combination Therapy in Wild-Type or NRTI-Resistant Strains of Diverse HIV-1 Subtypes. Viruses 2023; 15:1990. [PMID: 37896768 PMCID: PMC10612037 DOI: 10.3390/v15101990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Tenofovir disoproxil fumarate (TDF) and islatravir (ISL, 4'-ethynyl-2-fluoro-2'-deoxyadensine, or MK-8591) are highly potent nucleoside reverse transcriptase inhibitors. Resistance to TDF and ISL is conferred by K65R and M184V, respectively. Furthermore, K65R and M184V increase sensitivity to ISL and TDF, respectively. Therefore, these two nucleoside analogs have opposing resistance profiles and could present a high genetic barrier to resistance. To explore resistance to TDF and ISL in combination, we performed passaging experiments with HIV-1 WT, K65R, or M184V in the presence of ISL and TDF. We identified K65R, M184V, and S68G/N mutations. The mutant most resistant to ISL was S68N/M184V, yet it remained susceptible to TDF. To further confirm our cellular findings, we implemented an endogenous reverse transcriptase assay to verify in vitro potency. To better understand the impact of these resistance mutations in the context of global infection, we determined potency of ISL and TDF against HIV subtypes A, B, C, D, and circulating recombinant forms (CRF) 01_AE and 02_AG with and without resistance mutations. In all isolates studied, we found K65R imparted hypersensitivity to ISL whereas M184V conferred resistance. We demonstrated that the S68G polymorphism can enhance fitness of drug-resistant mutants in some genetic backgrounds. Collectively, the data suggest that the opposing resistance profiles of ISL and TDF suggest that a combination of the two drugs could be a promising drug regimen for the treatment of patients infected with any HIV-1 subtype, including those who have failed 3TC/FTC-based therapies.
Collapse
Affiliation(s)
- Maria E. Cilento
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xin Wen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Aaron B. Reeve
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Obiaara B. Ukah
- CS Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Alexa A. Snyder
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ciro M. Carrillo
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cole P. Smith
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kristin Edwards
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Claudia C. Wahoski
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Deborah R. Kitzler
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eiichi N. Kodama
- Division of Infectious Disease, International Institute of Disaster Science, Tohoku University, Sendai 980-8572, Japan
| | - Hiroaki Mitsuya
- Department of Refractory Viral Infections, National Center for Global Health & Medicine Research Institute, Tokyo 162-8655, Japan
- Experimental Retrovirology Section, HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Clinical Sciences, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | - Michael A. Parniak
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Philip R. Tedbury
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stefan G. Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
40
|
Jaha B, Schenkel CD, Jörimann L, Huber M, Zaheri M, Neumann K, Leemann C, Calmy A, Cavassini M, Kouyos RD, Günthard HF, Metzner KJ. Prevalence of HIV-1 drug resistance mutations in proviral DNA in the Swiss HIV Cohort Study, a retrospective study from 1995 to 2018. J Antimicrob Chemother 2023; 78:2323-2334. [PMID: 37545164 PMCID: PMC10477134 DOI: 10.1093/jac/dkad240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/19/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Genotypic resistance testing (GRT) is routinely performed upon diagnosis of HIV-1 infection or during virological failure using plasma viral RNA. An alternative source for GRT could be cellular HIV-1 DNA. OBJECTIVES A substantial number of participants in the Swiss HIV Cohort Study (SHCS) never received GRT. We applied a method that enables access to the near full-length proviral HIV-1 genome without requiring detectable viraemia. METHODS Nine hundred and sixty-two PBMC specimens were received. Our two-step nested PCR protocol was applied to generate two overlapping long-range amplicons of the HIV-1 genome, sequenced by next-generation sequencing (NGS) and analysed by MinVar, a pipeline to detect drug resistance mutations (DRMs). RESULTS Six hundred and eighty-one (70.8%) of the samples were successfully amplified, sequenced and analysed by MinVar. Only partial information of the pol gene was contained in 82/681 (12%), probably due to naturally occurring deletions in the proviral sequence. All common HIV-1 subtypes were successfully sequenced. We detected at least one major DRM at high frequency (≥15%) in 331/599 (55.3%) individuals. Excluding APOBEC-signature (G-to-A mutation) DRMs, 145/599 (24.2%) individuals carried at least one major DRM. RT-inhibitor DRMs were most prevalent. The experienced time on ART was significantly longer in DRM carriers (P = 0.001) independent of inclusion or exclusion of APOBEC-signature DRMs. CONCLUSIONS We successfully applied a reliable and efficient method to analyse near full-length HIV-1 proviral DNA and investigated DRMs in individuals with undetectable or low viraemia. Additionally, our data underscore the need for new computational tools to exclude APOBEC-related hypermutated NGS sequence reads for reporting DRMs.
Collapse
Affiliation(s)
- Bashkim Jaha
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Corinne D Schenkel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Lisa Jörimann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Maryam Zaheri
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Kathrin Neumann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Christine Leemann
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | - Matthias Cavassini
- Division of Infectious Diseases, University Hospital Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Roger D Kouyos
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Huldrych F Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Karin J Metzner
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
41
|
Ritsch M, Cassman NA, Saghaei S, Marz M. Navigating the Landscape: A Comprehensive Review of Current Virus Databases. Viruses 2023; 15:1834. [PMID: 37766241 PMCID: PMC10537806 DOI: 10.3390/v15091834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Viruses are abundant and diverse entities that have important roles in public health, ecology, and agriculture. The identification and surveillance of viruses rely on an understanding of their genome organization, sequences, and replication strategy. Despite technological advancements in sequencing methods, our current understanding of virus diversity remains incomplete, highlighting the need to explore undiscovered viruses. Virus databases play a crucial role in providing access to sequences, annotations and other metadata, and analysis tools for studying viruses. However, there has not been a comprehensive review of virus databases in the last five years. This study aimed to fill this gap by identifying 24 active virus databases and included an extensive evaluation of their content, functionality and compliance with the FAIR principles. In this study, we thoroughly assessed the search capabilities of five database catalogs, which serve as comprehensive repositories housing a diverse array of databases and offering essential metadata. Moreover, we conducted a comprehensive review of different types of errors, encompassing taxonomy, names, missing information, sequences, sequence orientation, and chimeric sequences, with the intention of empowering users to effectively tackle these challenges. We expect this review to aid users in selecting suitable virus databases and other resources, and to help databases in error management and improve their adherence to the FAIR principles. The databases listed here represent the current knowledge of viruses and will help aid users find databases of interest based on content, functionality, and scope. The use of virus databases is integral to gaining new insights into the biology, evolution, and transmission of viruses, and developing new strategies to manage virus outbreaks and preserve global health.
Collapse
Affiliation(s)
- Muriel Ritsch
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, 07743 Jena, Germany;
- European Virus Bioinformatics Center, 07743 Jena, Germany
| | - Noriko A. Cassman
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, 07743 Jena, Germany;
- European Virus Bioinformatics Center, 07743 Jena, Germany
| | - Shahram Saghaei
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, 07743 Jena, Germany;
- European Virus Bioinformatics Center, 07743 Jena, Germany
| | - Manja Marz
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, 07743 Jena, Germany;
- European Virus Bioinformatics Center, 07743 Jena, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, 04103 Leipzig, Germany
- FLI Leibniz Institute for Age Research, 07745 Jena, Germany
| |
Collapse
|
42
|
Siddiqui D, Badar U, Javaid M, Farooqui N, Shah SA, Iftikhar A, Sultan F, Mir F, Furqan S, Mahmood SF, Abidi SH. Genetic and antiretroviral drug resistance mutations analysis of reverse transcriptase and protease gene from Pakistani people living with HIV-1. PLoS One 2023; 18:e0290425. [PMID: 37616294 PMCID: PMC10449192 DOI: 10.1371/journal.pone.0290425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Antiretroviral therapy (ART) effectiveness is compromised by the emergence of HIV drug resistance mutations (DRM) and can lead to the failure of ART. Apart from intrinsic viral factors, non-compliance with drugs and/or the use of sub-optimum therapy can lead to the emergence of DRMs. In Pakistan HIV currently exists as a concentrated epidemic, however, ART coverage is very low, and drug adherence is poor. ART is selected assuming without baseline genotyping. Pakistan has recently seen a rise in treatment failures, but the country's actual burden of DRM is still unknown. In this study, we perform the genetic and drug resistance analysis of the pol gene from Pakistani HIV-positive ART-naïve and ART-experienced individuals. METHODS In this study, HIV-1 pol was sequenced from 146 HIV-1 positive individuals, divided into ART-naïve (n = 37) and ART-experienced (n = 109). The sequences were also used to determine HIV-1 subtypes, the prevalence of DRM, and pol genetic variability. RESULTS DRM analysis identified numerous DRMs against reverse transcriptase inhibitors in both ART-naïve and ART-experienced groups, including a few that are classified as rare. Additionally, the ART-experienced group showed mutations associated with resistance to protease inhibitors. Genetic analysis showed negative selection pressure in both groups, but a higher rate of evolution in the ART-naïve group. CONCLUSION High prevalence of DRMs, especially against previous first-line treatment in ART- naïve and the accumulation of DRMs in ART-experienced groups is concerning and warrants that a more extensive DRM survey be carried out to inform first-line and second-line ART regimen recommendations.
Collapse
Affiliation(s)
- Dilsha Siddiqui
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
- Department of Genetics, University of Karachi, Karachi, Pakistan
| | - Uzma Badar
- Department of Genetics, University of Karachi, Karachi, Pakistan
| | | | - Nida Farooqui
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Ayesha Iftikhar
- Shaukat Khanum Memorial Hospital and Research Centre, Lahore, Pakistan
| | - Faisal Sultan
- Shaukat Khanum Memorial Hospital and Research Centre, Lahore, Pakistan
| | - Fatima Mir
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sofia Furqan
- National AIDS Control Program, Ministry of Health, Islamabad, Pakistan
| | | | - Syed Hani Abidi
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, Kazakhstan
| |
Collapse
|
43
|
Kim MJ, Yu KL, Han R, Lee Y, Oh K, You JC. Identification of a Non-Nucleoside Reverse Transcriptase Inhibitor against Human Immunodeficiency Virus-1. ACS Infect Dis 2023; 9:1582-1592. [PMID: 37415514 DOI: 10.1021/acsinfecdis.3c00166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
The HIV-1 infection epidemic remains a global health problem. Current antiretroviral treatments are effective in controlling the progression of a severe infection. However, the emergence of drug resistance requires an urgent identification of new treatment regimes. HIV-1 reverse transcriptase (RTs) has been a successful therapeutic target owing to its high specificity and potent antiviral properties; therefore, it has become an essential component of current HIV-1 standard treatments. This study identified a new HIV-1 RTs inhibitor (Compound #8) that is structurally unique and greatly effective against HIV-1 through chemical library screening and a medicinal chemistry program by analyzing the structure-activity relationship (SAR). Further analysis of molecular docking and mechanisms of action demonstrated that Compound #8 is a novel type of HIV-1 non-nucleoside reverse transcriptase inhibitor (NNRTI) with a flexible binding mode. Therefore, it exhibits great therapeutic potential when combined with other existing HIV-1 drugs. Our current studies suggest that Compound #8 is a promising novel scaffold for the development of new HIV-1 treatments.
Collapse
Affiliation(s)
- Min-Jung Kim
- Avixgen Inc., 2477 Nambusunhwan-ro, Seocho, Seoul 06725, Republic of Korea
| | - Kyung Lee Yu
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho, Seoul 06591, Republic of Korea
| | - Ri Han
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea
| | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea
| | - Kyungsoo Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea
- Center for Metareceptome Research, Graduate School of Pharmaceutical Sciences, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Republic of Korea
| | - Ji Chang You
- Avixgen Inc., 2477 Nambusunhwan-ro, Seocho, Seoul 06725, Republic of Korea
- National Research Laboratory of Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho, Seoul 06591, Republic of Korea
| |
Collapse
|
44
|
Li M, Oliveira Passos D, Shan Z, Smith SJ, Sun Q, Biswas A, Choudhuri I, Strutzenberg TS, Haldane A, Deng N, Li Z, Zhao XZ, Briganti L, Kvaratskhelia M, Burke TR, Levy RM, Hughes SH, Craigie R, Lyumkis D. Mechanisms of HIV-1 integrase resistance to dolutegravir and potent inhibition of drug-resistant variants. SCIENCE ADVANCES 2023; 9:eadg5953. [PMID: 37478179 PMCID: PMC11803526 DOI: 10.1126/sciadv.adg5953] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/16/2023] [Indexed: 07/23/2023]
Abstract
HIV-1 infection depends on the integration of viral DNA into host chromatin. Integration is mediated by the viral enzyme integrase and is blocked by integrase strand transfer inhibitors (INSTIs), first-line antiretroviral therapeutics widely used in the clinic. Resistance to even the best INSTIs is a problem, and the mechanisms of resistance are poorly understood. Here, we analyze combinations of the mutations E138K, G140A/S, and Q148H/K/R, which confer resistance to INSTIs. The investigational drug 4d more effectively inhibited the mutants compared with the approved drug Dolutegravir (DTG). We present 11 new cryo-EM structures of drug-resistant HIV-1 intasomes bound to DTG or 4d, with better than 3-Å resolution. These structures, complemented with free energy simulations, virology, and enzymology, explain the mechanisms of DTG resistance involving E138K + G140A/S + Q148H/K/R and show why 4d maintains potency better than DTG. These data establish a foundation for further development of INSTIs that potently inhibit resistant forms in integrase.
Collapse
Affiliation(s)
- Min Li
- National Institute of Diabetes and Digestive Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Zelin Shan
- The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Steven J. Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Qinfang Sun
- Center for Biophysics and Computational Biology, and Department of Chemistry, Temple University, Philadelphia, PA 19122, USA
| | - Avik Biswas
- The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Center for Biophysics and Computational Biology and Department of Physics, Temple University, Philadelphia, PA 19122, USA
| | - Indrani Choudhuri
- Center for Biophysics and Computational Biology, and Department of Chemistry, Temple University, Philadelphia, PA 19122, USA
| | | | - Allan Haldane
- Center for Biophysics and Computational Biology and Department of Physics, Temple University, Philadelphia, PA 19122, USA
| | - Nanjie Deng
- Department of Chemistry and Physical Sciences, Pace University, New York, NY, 10038, USA
| | - Zhaoyang Li
- National Institute of Diabetes and Digestive Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xue Zhi Zhao
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Lorenzo Briganti
- Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mamuka Kvaratskhelia
- Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Terrence R. Burke
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Ronald M. Levy
- Center for Biophysics and Computational Biology and Department of Physics, Temple University, Philadelphia, PA 19122, USA
| | - Stephen H. Hughes
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Robert Craigie
- National Institute of Diabetes and Digestive Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dmitry Lyumkis
- The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Graduate School of Biological Sciences, Section of Molecular Biology, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
45
|
Novitsky V, Nyandiko W, Vreeman R, DeLong AK, Howison M, Manne A, Aluoch J, Chory A, Sang F, Ashimosi C, Jepkemboi E, Orido M, Hogan JW, Kantor R. Added Value of Next Generation Sequencing in Characterizing the Evolution of HIV-1 Drug Resistance in Kenyan Youth. Viruses 2023; 15:1416. [PMID: 37515104 PMCID: PMC10383797 DOI: 10.3390/v15071416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Drug resistance remains a global challenge in children and adolescents living with HIV (CALWH). Characterizing resistance evolution, specifically using next generation sequencing (NGS) can potentially inform care, but remains understudied, particularly in antiretroviral therapy (ART)-experienced CALWH in resource-limited settings. We conducted reverse-transcriptase NGS and investigated short-and long-term resistance evolution and its predicted impact in a well-characterized cohort of Kenyan CALWH failing 1st-line ART and followed for up to ~8 years. Drug resistance mutation (DRM) evolution types were determined by NGS frequency changes over time, defined as evolving (up-trending and crossing the 20% NGS threshold), reverting (down-trending and crossing the 20% threshold) or other. Exploratory analyses assessed potential impacts of minority resistance variants on evolution. Evolution was detected in 93% of 42 participants, including 91% of 22 with short-term follow-up, 100% of 7 with long-term follow-up without regimen change, and 95% of 19 with long-term follow-up with regimen change. Evolving DRMs were identified in 60% and minority resistance variants evolved in 17%, with exploratory analysis suggesting greater rate of evolution of minority resistance variants under drug selection pressure and higher predicted drug resistance scores in the presence of minority DRMs. Despite high-level pre-existing resistance, NGS-based longitudinal follow-up of this small but unique cohort of Kenyan CALWH demonstrated continued DRM evolution, at times including low-level DRMs detected only by NGS, with predicted impact on care. NGS can inform better understanding of DRM evolution and dynamics and possibly improve care. The clinical significance of these findings should be further evaluated.
Collapse
Affiliation(s)
- Vlad Novitsky
- Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Winstone Nyandiko
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
- College of Health Sciences, Moi University, Eldoret 30100, Kenya
| | - Rachel Vreeman
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
- Department of Global Health and Health System Design, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Arnhold Institute for Global Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Allison K DeLong
- School of Public Health, Brown University, Providence, RI 02912, USA
| | - Mark Howison
- Research Improving People's Lives, Providence, RI 02903, USA
| | - Akarsh Manne
- Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Josephine Aluoch
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
| | - Ashley Chory
- Department of Global Health and Health System Design, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Festus Sang
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
| | - Celestine Ashimosi
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
| | - Eslyne Jepkemboi
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
| | - Millicent Orido
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
| | - Joseph W Hogan
- Academic Model Providing Access to Healthcare (AMPATH), Eldoret 30100, Kenya
- School of Public Health, Brown University, Providence, RI 02912, USA
| | - Rami Kantor
- Alpert Medical School, Brown University, Providence, RI 02912, USA
| |
Collapse
|
46
|
Single-center experience evaluating and initiating people with HIV on long-acting cabotegravir/rilpivirine. AIDS 2023; 37:605-609. [PMID: 36730069 DOI: 10.1097/qad.0000000000003446] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To describe our experience evaluating and initiating individuals on long-acting injectable (LAI) cabotegravir/rilpivirine (CAB/RPV) and evaluate factors associated with starting LAI CAB/RPV and reasons for not starting. DESIGN We conducted a retrospective single-center study at the UC San Diego Owen Clinic. METHODS We included all individuals who expressed interest in treatment with LAI CAB/RPV between April 2021 and June 2022 who had a definitive decision made on starting LAI CAB/RPV. RESULTS In total, 383 individuals were included with 201 (52.5%) initiating LAI CAB/RPV. Those who initiated LAI CAB/RPV were younger ( P = 0.02) and were more likely to be on a two-drug regimen or first-generation integrase inhibitor regimen and less likely to be on a protease inhibitor or multiclass regimen. The most common reasons for not starting LAI CAB/RPV were inconsistent clinic attendance or difficulty being contacted and patient choice not to start. Of those who had a proviral DNA resistance test as workup for LAI CAB/RPV ( n = 135), 18.5% had a resistance mutation identified that may have impacted the activity of LAI CAB/RPV. CONCLUSION Despite novel challenges over half of our cohort initiated LAI CAB/RPV. Evaluating for potential non-nucleoside reverse transcriptase inhibitor resistance is an important part of the workup for LAI CAB/RPV and proviral DNA resistance testing can be an additional tool to identify potential resistance.
Collapse
|
47
|
Novitsky V, Steingrimsson J, Howison M, Dunn CW, Gillani FS, Fulton J, Bertrand T, Howe K, Bhattarai L, Ronquillo G, MacAskill M, Bandy U, Hogan J, Kantor R. Not all clusters are equal: dynamics of molecular HIV-1 clusters in a statewide Rhode Island epidemic. AIDS 2023; 37:389-399. [PMID: 36695355 PMCID: PMC9881752 DOI: 10.1097/qad.0000000000003426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Molecular epidemiology is a powerful tool to characterize HIV epidemics and prioritize public health interventions. Typically, HIV clusters are assumed to have uniform patterns over time. We hypothesized that assessment of cluster evolution would reveal distinct cluster behavior, possibly improving molecular epidemic characterization, towards disrupting HIV transmission. DESIGN Retrospective cohort. METHODS Annual phylogenies were inferred by cumulative aggregation of all available HIV-1 pol sequences of individuals with HIV-1 in Rhode Island (RI) between 1990 and 2020, representing a statewide epidemic. Molecular clusters were detected in annual phylogenies by strict and relaxed cluster definition criteria, and the impact of annual newly-diagnosed HIV-1 cases to the structure of individual clusters was examined over time. RESULTS Of 2153 individuals, 31% (strict criteria) - 47% (relaxed criteria) clustered. Longitudinal tracking of individual clusters identified three cluster types: normal, semi-normal and abnormal. Normal clusters (83-87% of all identified clusters) showed predicted growing/plateauing dynamics, with approximately three-fold higher growth rates in large (15-18%) vs. small (∼5%) clusters. Semi-normal clusters (1-2% of all clusters) temporarily fluctuated in size and composition. Abnormal clusters (11-16% of all clusters) demonstrated collapses and re-arrangements over time. Borderline values of cluster-defining parameters explained dynamics of non-normal clusters. CONCLUSIONS Comprehensive tracing of molecular HIV clusters over time in a statewide epidemic identified distinct cluster types, likely missed in cross-sectional analyses, demonstrating that not all clusters are equal. This knowledge challenges current perceptions of consistent cluster behavior over time and could improve molecular surveillance of local HIV epidemics to better inform public health strategies.
Collapse
Affiliation(s)
| | | | - Mark Howison
- Research Improving People’s Lives, Providence, RI, USA
| | | | | | | | | | | | | | | | | | - Utpala Bandy
- Rhode Island Department of Health, Providence, RI, USA
| | | | | |
Collapse
|
48
|
Mathew A, Ismael N, Meeds H, Vubil A, Zicai AF, Mabunda N, Blackard JT. Hepatitis B virus genotypes and drug resistance mutations circulating in blood donors in Beira, Mozambique. PLoS One 2023; 18:e0281855. [PMID: 36795797 PMCID: PMC9934330 DOI: 10.1371/journal.pone.0281855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Hepatitis B virus (HBV) infects nearly 300 million people and is the leading cause of hepatitis and hepatocellular carcinoma worldwide. Despite the high burden of HBV in sub-Saharan Africa, countries such as Mozambique have limited data available on circulating HBV genotypes and the presence of drug resistance mutations. Blood donors from Beira, Mozambique were tested for HBV surface antigen (HBsAg) and HBV DNA at the Instituto Nacional de Saúde in Maputo, Mozambique. Regardless of HBsAg status, donors with detectable HBV DNA were evaluated for HBV genotype. PCR was performed with primers amplifying a 2.1-2.2 kilobase fragment of the HBV genome. PCR products were submitted for next generation sequencing (NGS), and consensus sequences were evaluated for HBV genotype, recombination, and the presence or absence of drug resistance mutations. Of the 1281 blood donors tested, 74 had quantifiable HBV DNA. The polymerase gene could be amplified from 45 of 58 (77.6%) individuals with chronic HBV infection and 12 of 16 (75%) with occult HBV infection. Among these 57, 51 (89.5%) sequences belonged to HBV genotype A1, while 6 (10.5%) were HBV genotype E. All genotype E sequences were E/A recombinants, and clustered separately from other genotype E references. Genotype A samples had a median viral load of 637 IU/mL, while genotype E samples had a median viral load of 476,084 IU/mL. No drug resistance mutations were observed in the consensus sequences. The current study demonstrates the genotypic diversity of HBV in blood donors in Mozambique, but the absence of dominant (consensus) drug resistance mutations. Studies in other at-risk populations are essential for understanding the epidemiology, risk of liver disease, and likelihood of treatment resistance in resource-limited settings.
Collapse
Affiliation(s)
- Ann Mathew
- Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Nalia Ismael
- Instituto Nacional de Saúde, Marracuene, Mozambique
| | - Heidi Meeds
- Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Adolfo Vubil
- Instituto Nacional de Saúde, Marracuene, Mozambique
| | | | | | - Jason T. Blackard
- Division of Digestive Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| |
Collapse
|
49
|
Steytler J, van der Ryst E, Craig C, Van Baelen B, Nuttall J, van Niekerk N, Mellors J, Parikh U, Wallis C, for the IPM 007 Study Team. Clinical Presentation, Treatment Response, and Virology Outcomes of Women Who Seroconverted in the Dapivirine Vaginal Ring Trials-The Ring Study and DREAM. Clin Infect Dis 2023; 76:389-397. [PMID: 36189636 PMCID: PMC10169386 DOI: 10.1093/cid/ciac804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Participants with human immunodeficiency virus (HIV) seroconversion in The Ring Study, a phase 3 trial of dapivirine vaginal ring (DVR), or in the open-label extension trial dapivirine ring extended access and monitoring (DREAM) were offered enrollment in an observational cohort study (IPM 007) to assess clinical presentation and response to antiretroviral therapy (ART). METHODS Participants' HIV infection was managed at local treatment clinics according to national treatment guidelines. IPM 007 study visits occurred 3 and 6 months after enrollment and every 6 months thereafter. Assessments included plasma HIV-1 RNA, CD4+ T-cell counts, and recording of HIV/AIDS-associated events and antiretroviral use. Post hoc virology analyses were performed for participants identified with virologic failure. RESULTS One hundred fifty-one of 179 eligible participants (84.4%) enrolled into IPM 007; 103 had previously received the DVR in the Ring or DREAM studies, and 48 had received placebo in The Ring Study. HIV-1 RNA and CD4+ T-cell counts after 12 months' follow-up were similar for participants who used the DVR in The Ring Study and DREAM, compared to those who received placebo. Of the 78 participants with a study visit approximately 6 months after ART initiation, 59 (75.6%) had HIV-1 RNA <40 copies/mL (The Ring Study: placebo: 13/23 [56.5%]; DVR: 32/39 [82.1%]; DREAM [DVR]: 14/16 [87.5%]). Post hoc virology analysis indicated that genotypic patterns observed at virologic failure were as expected of a nonnucleoside reverse transcriptase inhibitor (NNRTI)-based regimen. CONCLUSIONS Seroconversion during DVR use did not negatively affect clinical presentation or treatment outcome. Mutation patterns at virologic failure were in line with individuals failing an NNRTI-based regimen. CLINICAL TRIALS REGISTRATION NCT01618058.
Collapse
Affiliation(s)
- John Steytler
- International Partnership for Microbicides South Africa NPC, Johannesburg, South Africa
| | | | - Charles Craig
- Research Virology Consulting Ltd, Cambridgeshire, United Kingdom
| | | | - Jeremy Nuttall
- International Partnership for Microbicides, Silver Spring, Maryland, USA
| | - Neliëtte van Niekerk
- International Partnership for Microbicides South Africa NPC, Johannesburg, South Africa
| | - John Mellors
- Microbicide Trials Network Virology Core Laboratory, University of Pittsburgh, Pennsylvania, USA
| | - Urvi Parikh
- Microbicide Trials Network Virology Core Laboratory, University of Pittsburgh, Pennsylvania, USA
| | - Carole Wallis
- Bio-Analytical Research Corporation Laboratory, Johannesburg, South Africa
| | | |
Collapse
|
50
|
Switching to coformulated bictegravir, emtricitabine, and tenofovir alafenamide maintained viral suppression in adults with historical virological failures and K65N/R mutation. Int J Infect Dis 2023; 126:39-47. [PMID: 36384186 DOI: 10.1016/j.ijid.2022.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Real-world experience with coformulated bictegravir, emtricitabine, and tenofovir alafenamide (BIC/FTC/TAF) is sparse as a switch regimen among people living with HIV (PLWH) having achieved viral suppression after previous virologic failures with the emergence of K65N/R. METHODS In this retrospective study, PLWH aged ≥20 years who had previous virologic failures with emergent K65N/R were included for switching to BIC/FTC/TAF after having achieved plasma HIV RNA load (PVL) <200 copies/ml for ≥3 months. PLWH were excluded if integrase inhibitor resistance-associated mutations were detected. The primary end point was losing virologic control (PVL >50 copies/ml) at week 48 using a modified US Food and Drug Administration snapshot algorithm. RESULTS A total of 72 PLWH with K65N/R who switched to BIC/FTC/TAF were identified. A total of 42 (59.7%) had concurrent M184V/I, and 9 (12.5%) had ≥1 thymidine analog mutations. The median duration of viral suppression was 4.7 years (interquartile range 2.3-5.8), and 97.2% (n = 70) had PVL <50 copies/ml before switching. After a median observation of 98.6 weeks (interquartile range 77.9-120.3), 94.4% (n = 68) continued BIC/FTC/TAF. At week 48, the rate of losing virologic control was 2.8% (2/72). M184V/I was not associated with viral rebound. CONCLUSION Despite the emergence of K65N/R +/- M184V/I after virologic failures, BIC/FTC/TAF could be an option for simplification after viral suppression.
Collapse
|