1
|
Puthanakit T, Aurpibul L, Lopez M, Wang M, Ciuffa M, Bontempo G, Cheung SYA, Deprez I, Buchanan AM, Vavro C, McKenna M, Min S, Tan LK. Efficacy and Safety of the Two-Drug Regimen Dolutegravir-Lamivudine in Adolescents Living With HIV-1 Naive to Antiretroviral Therapy at 48 Weeks (DANCE): A Single-Arm, Open-Label, Phase 3b Trial. J Acquir Immune Defic Syndr 2025; 99:202-210. [PMID: 39988749 DOI: 10.1097/qai.0000000000003655] [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: 07/26/2024] [Accepted: 01/07/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Dolutegravir-lamivudine is recommended for initial antiretroviral therapy (ART) in adults living with HIV-1; however, no clinical trials have assessed this regimen in adolescents, a potentially more challenging population to treat. We evaluated efficacy, safety, and pharmacokinetics of dolutegravir-lamivudine as initial ART in adolescents living with HIV-1. SETTING Nine centers in Thailand, Kenya, and South Africa. METHODS In the single-arm, open-label, phase 3b DANCE study (NCT03682848), adolescents naive to ART aged ≥12 to <18 years, weighing ≥25 kg, with plasma HIV-1 RNA 1000 to ≤500,000 copies/mL received single once-daily dolutegravir-lamivudine fixed-dose combination tablet orally for 48 weeks (treatment phase). The primary endpoint was proportion of participants with HIV-1 RNA <50 copies/mL at Week 48 [Snapshot; intention-to-treat-exposed (ITT-E) population]. Safety outcomes were evaluated in the safety population. Both the ITT-E and safety populations consisted of participants who received ≥1 dose of dolutegravir-lamivudine (N = 32). A sensitivity analysis was performed using data from all but 2 participants who were withdrawn before Week 48 because of site closure (n = 30). RESULTS At Week 48, 26 of 32 (81%; 95% CI: 64 to 93) participants had HIV-1 RNA <50 copies/mL; in sensitivity analyses, 26 of 30 (87%; 95% CI: 69 to 96) participants achieved virologic suppression. No confirmed virologic withdrawals or deaths were reported. One drug-related adverse event was reported (grade 3 decreased glomerular filtration rate) and was the only adverse event leading to study withdrawal at data cut. Pharmacokinetic parameters were comparable with adult systemic exposure ranges. CONCLUSIONS Dolutegravir-lamivudine demonstrated efficacy and safety as initial ART in adolescents with HIV-1 through 48 weeks.
Collapse
Affiliation(s)
| | - Linda Aurpibul
- Research Institute for Health Sciences (RIHES), Chiang Mai University, Chiang Mai, Thailand
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Phiri K, Hagstrom C, Lungu E, Balakasi K, Songo J, Makwaya A, Smith D, Worku A, Hoffman R, Phiri S, Dovel K, van Oosterhout JJ. Barriers to viral suppression in children aged 9 years or younger on dolutegravir-based antiretroviral therapy in Malawi, a mixed-methods study. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004510. [PMID: 40445885 PMCID: PMC12124501 DOI: 10.1371/journal.pgph.0004510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 04/28/2025] [Indexed: 06/02/2025]
Abstract
After the transition to pediatric dolutegravir-based regimens, viral load (VL) suppression among children with HIV (CWH) in Malawi has remained suboptimal. This mixed-methods study assessed factors associated with high VL among young CWH on dolutegravir-based antiretroviral therapy (ART) and explored adherence barriers from primary-caregiver and healthcare worker perspectives. Between April-July 2023, we performed an unmatched case-control study at 49 Malawian health facilities. We included CWH aged ≤9 years, on dolutegravir-based ART, with a routine VL test-result that was high (≥1,000 copies/mL) for cases, or suppressed (<200 copies/mL) for controls. Using mixed-effect modified Poisson regression, we determined factors associated with high VL, adjusting for sex, site and district. To assess adherence barriers, we conducted in-depth interviews (IDIs) with randomly selected caregivers of CWH with high VL and with healthcare workers providing pediatric HIV care. Data were analyzed using a hybrid thematic approach that combined deductive and inductive coding strategies. We enrolled 538 CWH: 222 cases, with high VL and 316 controls, with suppressed VL. Duration on ART > 4 years (aRR = 0.86, 95% CI: 0.77-0.95) and ≥2 interruption in treatment episodes (≥28 days late for clinic appointment) in the 12 months before VL sample collection (aRR = 1.47, 95% CI: 1.28-1.68) were significantly associated with high VL. Through 54 IDIs (30-caregivers, 24-healthcare workers), five key adherence challenges were identified and affected the children ≤5 years the most: resistance to daily medication, difficulties taking multiple pills, food insecurity, fear of unintentional disclosure, and inability to attend clinic appointments consistently. This study highlights that behavioral, socio-economic and psychosocial factors influences ART adherence among CWH. Duration on ART and recent interruptions in treatment were associated with high VL, stressing the need for targeted interventions that will require health-system and client-level approaches to improve VL suppression among CWH in Malawi and similar settings.
Collapse
Affiliation(s)
| | - Christine Hagstrom
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | | | | | | | | | - Deanna Smith
- Partners in Hope, Lilongwe, Malawi
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Anteneh Worku
- Department of Health, Nutrition and Population, USAID Malawi, Lilongwe, Malawi
| | - Risa Hoffman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Sam Phiri
- Partners in Hope, Lilongwe, Malawi
- School of Global and Public Health, Kamuzu University of Health Sciences, Lilongwe, Malawi
| | - Kathryn Dovel
- Partners in Hope, Lilongwe, Malawi
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Joep J. van Oosterhout
- Partners in Hope, Lilongwe, Malawi
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| |
Collapse
|
3
|
Dollfus C, Le Chenadec J, Reliquet V, Alby-Laurent F, Brehin C, Caseris M, Runel-Belliard C, Blanche S, Warszawski J, Faye A, Frange P. Outcomes and challenges in adolescents with HIV in France: a nationwide cohort study over 35 years. AIDS 2025; 39:667-675. [PMID: 39813288 DOI: 10.1097/qad.0000000000004121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025]
Abstract
OBJECTIVE Most data published on adolescents with HIV (AWH) have been collected before the large diffusion of second-generation integrase strand transfer inhibitors (INSTI) among the pediatric population. We analyzed the nationwide ANRS-MIE CO10 Pediatric cohort to assess the changes over time in health and social outcomes of French AWH. DESIGN The cohort enrolled children born in France since 1985 and, from 2005, children diagnosed with HIV at ≤13 years, including those born abroad if antiretroviral-naive at first medical care in France. METHODS Adolescents aged ≥10 years at their last visit were included in this analysis. Their characteristics were compared between three periods of birth (1985-1993, 1994-1999, 2000-2010). RESULTS Overall, 529 AWH were included. Their median age at first HAART initiation decreased from 94 to 29 months ( P < 0.0001). At the last evaluation, the proportions of AWH receiving HAART, receiving INSTI, having HIV-RNA < 50 copies/mL and having CD4 count ≥500/μl increased over time ( P < 0.0001), reaching 98·7%, 53.3%, 81.3% and 85·0%, respectively, for those born in 2000-2010. The proportion of maternal and paternal orphans decreased until 14.4% and 11.0%, respectively, for AWH born recently. Compared to middle adolescents (15-17 years) born in 1994-1999, those born in 2000-2010 demonstrated higher academic success (69·2% versus 42·3%) and less frequent academic failure (4.6% versus 6.2%). CONCLUSIONS Despite spectacular improvement in their health and immunovirological status, AWH remain vulnerable compared to other French adolescents, with a higher risk of being orphan and/or experiencing academic failure. Specific interventions are required to improve their global quality of life.
Collapse
Affiliation(s)
- Catherine Dollfus
- Pediatric Hematology and Oncology Department, Trousseau Hospital, GHU Assistance Publique - Hôpitaux de Paris (APHP).Sorbonne Université, Paris
| | | | - Véronique Reliquet
- Infectious Diseases Department, CHU Hôtel Dieu and INSERM UIC 1413, Nantes University, Nantes
| | - Fanny Alby-Laurent
- Pediatric Hematology and Oncology Department, Trousseau Hospital, GHU Assistance Publique - Hôpitaux de Paris (APHP).Sorbonne Université, Paris
| | - Camille Brehin
- Paediatric Infectious Diseases Department, Children's Hospital, Toulouse University Hospital, Toulouse
| | - Marion Caseris
- Department of General Pediatrics and infectious diseases, Robert Debré Hospital, GHU APHP. Nord - Université Paris Cité, Paris
| | | | - Stéphane Blanche
- Pediatric Hematology and Immunology Department, Necker Hospital, GHU APHP. Centre - Université de Paris, Paris
| | - Josiane Warszawski
- INSERM CESP U1018, Université Paris-Saclay, Le Kremlin-Bicêtre
- Epidemiology and Public Health Department, GHU APHP.Université Paris-Saclay, Le Kremlin-Bicêtre
| | - Albert Faye
- Department of General Pediatrics and infectious diseases, Robert Debré Hospital, GHU APHP. Nord - Université Paris Cité, Paris
| | - Pierre Frange
- Pediatric Hematology and Immunology Department, Necker Hospital, GHU APHP. Centre - Université de Paris, Paris
- Laboratory of Clinical Microbiology, Necker Hospital, GHU APHP. Centre - Université de Paris
- URP 7328 FETUS, Université Paris Cité, Paris, France
| |
Collapse
|
4
|
White E, Kityo C, Spyer MJ, Mujuru HA, Nankya I, Kekitiinwa AR, Lugemwa A, Kaudha E, Liberty A, Cassim H, Archary M, Cotton MF, Ahimbisibwe GM, Cressey TR, Ngampiyaskul C, Srirompotong U, Behuhuma O, Saidi Y, Bamford A, Kobbe R, Nastouli E, Rojo P, Giaquinto C, Gibb DM, Ford D, Turkova A. Virological outcomes and genotypic resistance on dolutegravir-based antiretroviral therapy versus standard of care in children and adolescents: a secondary analysis of the ODYSSEY trial. Lancet HIV 2025; 12:e201-e213. [PMID: 39978387 DOI: 10.1016/s2352-3018(24)00155-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 02/22/2025]
Abstract
BACKGROUND ODYSSEY showed superior efficacy for dolutegravir-based antiretroviral therapy (ART) versus standard of care (SOC) in children living with HIV starting first-line or second-line ART aged 4 weeks or older. Here, we aim to compare virological outcomes and resistance in the dolutegravir group versus SOC for first-line and second-line ART up to 96 weeks. METHODS ODYSSEY was an open-label, multicentre, randomised, non-inferiority trial done in 29 centres in seven countries (Germany, Spain, South Africa, Thailand, the UK, Uganda, and Zimbabwe). ODYSSEY recruited children living with HIV aged at least 28 days and younger than 18 years, weighing at least 3 kg, starting first-line ART (ODYSSEY A), or switching to second-line therapy after treatment failure (ODYSSEY B). Children were randomly assigned (1:1) to dolutegravir plus two nucleoside or nucleotide reverse transcriptase inhibitors (NRTIs; dolutegravir group) versus the SOC group (non-nucleoside reverse transcriptase inhibitor [NNRTI], boosted protease inhibitor, or non-dolutegravir integrase strand-transfer inhibitor, plus two NRTIs). Two randomised cohorts were combined in this exploratory analysis: children weighing at least 14 kg were enrolled between Sept 20, 2016, and June 22, 2018, and children weighing less than 14 kg were enrolled between July 5, 2018, and Aug 26, 2019. Virological failure was defined as an inadequate virological response at week 24 with an ART switch or confirmed HIV-1 RNA viral load of at least 400 copies per mL after week 36. Virological suppression was defined as two consecutive viral loads of less than 400 copies per mL and was compared between groups, including an ART switch and death as competing risks. Children with virological failure were tested for post-failure genotypic resistance, with baseline results used to identify emergent resistance. Development of emergent resistance was a secondary trial outcome and all other outcomes are exploratory. ODYSSEY was registered with ClinicalTrials.gov (NCT02259127), EUDRACT (2014-002632-14), and ISRCTN (ISRCTN91737921). FINDINGS In ODYSSEY at enrolment, 381 participants started first-line ART (ODYSSEY A: 189 in the dolutegravir group and 192 in the SOC group) and 407 participants started second-line ART (ODYSSEY B: 202 in the dolutegravir group and 205 in the SOC group). 72 participants in ODYSSEY A and 13 participants in ODYSSEY B weighed less than 14 kg. 401 (51%) of 788 participants were female and 387 (49%) were male. Virological suppression occurred significantly earlier in the dolutegravir group (adjusted [cause-specific] hazard ratio [HR] 1·57 [95% CI 1·35 to 1·83]; p<0·0001). Overall, 51 (13%) participants had virological failure by 96 weeks in the dolutegravir group versus 86 (22%) in the SOC group (including 18 [10%] vs 43 [22%] in ODYSSEY A and in 33 [16%] vs 43 [21%] in ODYSSEY B; adjusted HR 0·56 [0·40 to 0·79]; p=0·0011). Among ODYSSEY B participants starting dolutegravir, virological failure was higher in children starting zidovudine (HR 2·22 [1·01 to 4·88]; p=0·048) and similar in those starting tenofovir disoproxil fumarate (1·19 [0·50 to 2·83]; p=0·70) compared with abacavir. Time to virological suppression was marginally faster in participants receiving second-line dolutegravir and abacavir with high-level abacavir resistance at baseline compared with those with no, low-level, intermediate-level resistance (cause-specific HR 1·70 [1·01 to 2·85]; p=0·046); and failure rates by week 96 were similar (HR 0·90 [0·23 to 3·61]; p=0·88). An estimated 1% (95% CI 0 to 2) of participants in the dolutegravir group versus 20% (14 to 26) in the SOC group in ODYSSEY A had emergent resistance to at least one drug-class within their first-line regimen (risk difference -20% [-25 to -14]; p<0·0001); 4% (1 to 6) versus 5% (2 to 8) had resistance to drug within their initial second-line regimen (risk difference -1% [-5 to 3]; p=0·60). 3% (0 to 5) of participants in the dolutegravir group had emergent integrase strand-transfer inhibitors resistance compared with 3% (1 to 6) of participants in the SOC group who had emergent resistance to the anchor drug (risk difference 0% [-4 to 3]; p=0·78). INTERPRETATION Dolutegravir led to faster virological suppression and lower risk of virological failure than NNRTIs and boosted protease inhibitor-based SOC. Participants starting second-line dolutegravir-based ART with an abacavir or tenofovir backbone were at lower risk of virological failure than those starting zidovudine. During first-line therapy, dolutegravir protected against emergent resistance; starting second-line therapy, the risk of emergent resistance to nucleoside reverse transcriptase inhibitor backbone, and anchor drugs, was similar among participants starting dolutegravir within their second-line regimen and those starting mainly boosted protease inhibitor-based SOC. FUNDING Penta Foundation, ViiV Healthcare, and UK Medical Research Council.
Collapse
Affiliation(s)
- Ellen White
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK.
| | - Cissy Kityo
- Joint Clinical Research Centre, Kampala, Uganda
| | - Moira J Spyer
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College London, London, UK; Hospitals NHS Trust, Advanced Pathogen Diagnostics Unit, University College London, London, UK
| | | | | | | | | | | | - Afaaf Liberty
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Haseena Cassim
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Moherndran Archary
- Department of Paediatrics and Child Health, Enhancing Care Foundation, King Edward VIII Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Mark F Cotton
- Department of Paediatrics and Child Health, Family Center for Research with Ubuntu, Stellenbosch University, Cape Town, South Africa
| | | | - Tim R Cressey
- Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | | | | | - Osee Behuhuma
- Africa Health Research Institute, Kwazulu-Natal, Durban, South Africa
| | - Yacine Saidi
- INSERM/ANRS SC10-US19, Essais thérapeutiques et maladies infectieuses, Villejuif, France
| | - Alasdair Bamford
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK; Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Robin Kobbe
- Institute for Infection Research and Vaccine Development, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Eleni Nastouli
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College London, London, UK; Hospitals NHS Trust, Advanced Pathogen Diagnostics Unit, University College London, London, UK
| | - Pablo Rojo
- Pediatric Infectious Diseases Unit, Hospital 12 de Octubre, Madrid, Spain
| | - Carlo Giaquinto
- Department of Women and Child Health, University of Padova, Padova, Italy; Fondazione Penta ETS, Padova, Italy
| | - Diana M Gibb
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Deborah Ford
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Anna Turkova
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK; Department of Paediatric Infectious Diseases, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
5
|
Konu YR, Takassi E, Peytavin G, Dapam N, Damond F, Oumarou WA, Zaidi M, Franco-Yusti AM, Dagnra CA, Le Hingrat Q, Coppée R, Descamps D, Diallo FBT, Ekouevi DK, Charpentier C. Pharmaco-virological Outcomes and Genotypic Resistance Profiles Among Children and Adolescents Receiving a Dolutegravir-Based Regimen in Togo. Clin Infect Dis 2025; 80:144-152. [PMID: 38748464 DOI: 10.1093/cid/ciae278] [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: 12/18/2023] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Few data are available on the real-world efficacy of receiving tenofovir-lamivudine-dolutegravir (-DTG) as human immunodeficiencyvirus (HIV) treatment, particularly among young people in West Africa. Here, we evaluated pharmaco-virological outcomes and resistance profiles among Togolese children and adolescents. METHODS A cross-sectional study was conducted in Lomé, Togo, enrolling antiretroviral-treated people with HIV aged from 18 months to 24 years. Plasma HIV-1 viral load and antiretroviral concentrations were measured. Next-generation sequencing of protease, reverse transcriptase (RT), and integrase was performed on all samples with viral loads >200 copies/mL. Drug resistance mutations (DRMs) were identified and interpreted using the ANRS-MIE algorithm. RESULTS 264 participants were enrolled (median age, 17 years); 226 received a DTG-based regimen for a median of 20.5 months. Among them, there was virological suppression at the 200-copies/mL threshold in 80.0% of the participants. Plasma DTG concentrations were adequate (ie, >640 ng/mL), suboptimal, and below the limit of quantification in 74.1%, 6.7%, and 19.2% of participants receiving DTG, respectively. Overall, viruses resistant to any of nucleoside RT inhibitors, non-NRTIs, and protease inhibitors were found in 52%, 66%, and 1.6% of participants, respectively. A major integrase inhibitor DRM was observed in 9.4% (n = 3/32; R263K, E138A-G140A-Q148R, and N155H) of participants with a viral load >200 copies/mL. CONCLUSIONS These first findings in a large series of adolescents in a low-income country showed a good virological response of 80% and the presence of an integrase DRM in 9.4% of virological failures, supporting the need to monitor DTG drug resistance to reduce the risk of resistance acquisition.
Collapse
Affiliation(s)
- Yao Rodion Konu
- Faculté des Sciences de la Santé, Département de Santé Publique, Université de Lomé, Lomé, Togo
- Centre Africain de Recherche en Epidémiologie et en Santé Publique (CARESP), Lomé, Togo
| | - Elom Takassi
- Service de Pédiatrie, CHU Sylvanus Olympio, Université de Lomé, Lomé, Togo
| | - Gilles Peytavin
- Service de Pharmacologie, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM), Infections, Antimicrobials, Modeling, Evolution (IAME), UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | | | - Florence Damond
- Service de Virologie, Université Paris Cité, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Wone Adama Oumarou
- Centre Africain de Recherche en Epidémiologie et en Santé Publique (CARESP), Lomé, Togo
| | - Meryem Zaidi
- Service de Virologie, Université Paris Cité, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Anna-Maria Franco-Yusti
- Service de Virologie, Université Paris Cité, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Claver A Dagnra
- Centre de Biologie Moléculaire et d'Immunologie, Université de Lomé, Lomé, Togo
| | - Quentin Le Hingrat
- Service de Virologie, Université Paris Cité, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Romain Coppée
- Service de Virologie, Université Paris Cité, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Diane Descamps
- Service de Virologie, Université Paris Cité, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | | | - Didier K Ekouevi
- Centre Africain de Recherche en Epidémiologie et en Santé Publique (CARESP), Lomé, Togo
- Faculty of Health Sciences, Department of Public Health, University of Lomé, Center for Training and Research in Public Health, Lomé, Togo
- Research Institute for Sustainable Development (IRD), University of Bordeaux, INSERM, Bordeaux Population Health Centre, UMR 1219, Bordeaux, France
| | - Charlotte Charpentier
- Service de Virologie, Université Paris Cité, INSERM, IAME, UMR 1137, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| |
Collapse
|
6
|
Njom Nlend AE, Fokam J, Mekoui S, Epée Ngoué J, Nga Motaze AC, Colizzi V, Perno CF, Ndjolo A, Halle-Ekane GE, Sandie A. Outcomes of HIV-infected children on antiretroviral therapy for at least 10 years at the Essos Hospital Centre, Cameroon: contributing to the elimination of paediatric AIDS in tropical settings. J Trop Pediatr 2024; 71:fmae052. [PMID: 39673741 DOI: 10.1093/tropej/fmae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2024]
Abstract
Evidence on long-term outcomes of children receiving antiretroviral therapy (ART) in low- and middle-income countries (LMICs) is of utmost importance to optimize current and future therapeutic strategies for HIV. We sought to ascertain the long-term responses among ART-experienced children and their potential implications. A retrospective, observational, facility-based cohort study was conducted among 136 ART-experienced children monitored for 10 years (2007-2017) at the Essos Hospital Centre in Yaoundé, Cameroon. Primary outcomes were good clinical response defined as the World Health Organization (WHO) clinical stage 1/2, immune restoration as CD4 >500 cells/mm3, viral suppression (VS) as viral load (VL) <1000, or viral undetectability as VL <40 copies/ml at the last follow-up and their predictors (P < .05). At ART initiation among the 90 eligible children, median (interquartile range) age was 29.5 (11-60) months, 53.3% were males, 34.5% were at WHO clinical stage 1/2, median-CD4 was 497 cells/mm3, and initial ART-regimens were mainly zidovudine-lamivudine-nevirapine (85.5%) and zidovudine-lamivudine-efavirenz (12.2%). After 10 years of follow-up, 36.7% were switched to protease-based ART regimens, those at WHO clinical stage 1/2 improved to 75.5%; median-CD4 increased to 854 cells/mm3; 83.3% achieved VS, while only 10% achieved viral undetectability. Following multivariate analysis, no predictor of VS or immune recovery was identified (P > .05). In this LMIC, long-term outcomes of children on ART appear encouraging, characterized by moderate VS rate and immune recovery. However, challenges in achieving viral undetectability indicate continuous viral replication and risks of drug resistance emergence that may jeopardize effectiveness of future paediatric ART strategies.
Collapse
Affiliation(s)
- Anne Esther Njom Nlend
- Departement of Paediatrics, National Social Welfare Hospital, Essos, Yaoundé, 99322, Cameroon
- Departement of Clinical Sciences, Higher Institute of Medical Technology, University of Douala, Yaoundé, 99322, Cameroon
- Research Department, Health Ebene Consulting, Yaoundé, 99322, Cameroon
| | - Joseph Fokam
- Virology Laboratory, Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management (CIRCB), Yaoundé, 99322, Cameroon
- Department of Medical Laboratory Science, Faculty of Health Sciences, University of Buea, Buea, 99322, Cameroon
- Department of Pediatrics, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, 99322, Cameroon
- National HIV Drug Resistance Prevention and Surveillance Working Group, Ministry of Public Health, Yaounde, 99322, Cameroon
- Permanent Secretariat, Central Technical Group, National AIDS Control Committee, Ministry of Public Health, Yaounde, 99322, Cameroon
| | - Suzanne Mekoui
- Departement of Clinical Sciences, Higher Institute of Medical Technology, University of Douala, Yaoundé, 99322, Cameroon
| | - Jeannette Epée Ngoué
- Department of Pediatrics, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, 99322, Cameroon
| | - Annie Carole Nga Motaze
- Departement of Paediatrics, National Social Welfare Hospital, Essos, Yaoundé, 99322, Cameroon
| | - Vittorio Colizzi
- Virology Laboratory, Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management (CIRCB), Yaoundé, 99322, Cameroon
- EUROPBIOPARK and UNESCO Board of Multidisciplinary Biotechnology, University of Rome Tor Vergata, Rome, 00045, Italy
- Faculty of Science and Technology, Evangelic University of Cameroon, Bandjoun, 99322, Cameroon
| | - Carlo-Federico Perno
- Virology Laboratory, Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management (CIRCB), Yaoundé, 99322, Cameroon
- Bambino Gèsu Pediatric Hospital, Rome, 00045, Italy
| | - Alexis Ndjolo
- Virology Laboratory, Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management (CIRCB), Yaoundé, 99322, Cameroon
- Department of Pediatrics, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, 99322, Cameroon
| | - Gregory-Edie Halle-Ekane
- Department of Medical Laboratory Science, Faculty of Health Sciences, University of Buea, Buea, 99322, Cameroon
| | - Arsene Sandie
- African Population and Health Research Centre, Dakar, Dakar 12500, Senegal
| |
Collapse
|
7
|
LaNoce E, Zhang DY, Garcia-Epelboim A, Su Y, Sun Y, Alepa G, Angelucci AR, Akay-Espinoza C, Jordan-Sciutto KL, Song H, Ming GL, Christian KM. Exposure to the antiretroviral drug dolutegravir impairs structure and neurogenesis in a forebrain organoid model of human embryonic cortical development. Front Mol Neurosci 2024; 17:1459877. [PMID: 39569018 PMCID: PMC11576471 DOI: 10.3389/fnmol.2024.1459877] [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: 07/05/2024] [Accepted: 09/13/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction For many therapeutic drugs, including antiretroviral drugs used to treat people living with HIV-1 (PLWH), we have little data on the potential effects on the developing human brain due to limited access to tissue and historical constraints on the inclusion of pregnant populations in clinical trials. Human induced pluripotent stem cells (iPSCs) offer a new avenue to gain insight on how drugs may impact human cell types representative of the developing central nervous system. To prevent vertical transmission of HIV and promote the health of pregnant PLWH, antiretroviral therapy must be initiated and/or maintained throughout pregnancy. However, many antiretroviral drugs are approved for widespread use following clinical testing only in non-pregnant populations and there may be limited information on potential teratogenicity until pregnancy outcomes are evaluated. The integrase strand transfer inhibitor dolutegravir (DTG) is a frontline antiretroviral drug that is effective in viral suppression of HIV but was previously reported to be associated with a slight increase in the risk for neural tube defects in one study, although this has not been replicated in other cohorts. Methods To directly investigate the potential impact of DTG on human cortical neurogenesis, we measured the effects of daily drug exposure on the early stages of corticogenesis in a human iPSC-based forebrain organoid model. We quantified organoid size and structure and analyzed gene and protein expression to evaluate the impact of several doses of DTG on organoid development. Results We observed deficits in organoid structure and impaired neurogenesis in DTG-treated organoids compared to vehicle-treated control organoids after 20 or 40 days in culture. Our highest dose of DTG (10 μM) resulted in significantly smaller organoids with a reduced density of neural rosette structures compared to vehicle-treated controls. Mechanistically, RNA-sequencing and immunohistological analysis suggests dysregulated amino acid transport and activation of the integrated stress response in the DTG-treated organoids, and functionally, a small molecule integrated stress response inhibitor (ISRIB) could partially rescue increased expression of proteins related to cell cycle regulation. Discussion Together, these results illustrate the potential for human iPSC-based strategies to reveal biological processes during neurogenesis that may be affected by therapeutic drugs and provide complementary data in relevant human cell types to augment preclinical investigations of drug safety during pregnancy.
Collapse
Affiliation(s)
- Emma LaNoce
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y. Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alan Garcia-Epelboim
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yusha Sun
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Giana Alepa
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Angelina R. Angelucci
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cagla Akay-Espinoza
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kelly L. Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kimberly M. Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
8
|
Bishop MD, Korutaro V, Boyce CL, Beck IA, Styrchak SM, Knowles K, Ziemba L, Brummel SS, Coletti A, Jean-Philippe P, Chakhtoura N, Vhembo T, Cassim H, Owor M, Fairlie L, Moyo S, Chinula L, Lockman S, Frenkel LM. Characterizing HIV drug resistance in cases of vertical transmission in the VESTED randomized antiretroviral treatment trial. J Acquir Immune Defic Syndr 2024; 96:385-392. [PMID: 39175843 PMCID: PMC11338623 DOI: 10.1097/qai.0000000000003435] [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: 08/22/2023] [Accepted: 03/04/2024] [Indexed: 08/24/2024]
Abstract
Introduction VESTED (NCT03048422) compared the safety and efficacy of three antiretroviral treatment (ART) regimens in pregnant and postpartum women: dolutegravir+emtricitabine/tenofovir alafenamide fumarate; dolutegravir+emtricitabine/tenofovir disoproxil fumarate (TDF); efavirenz/emtricitabine/TDF. Vertical HIV transmission (VT) occurred to 4/617 (0.60%) live-born infants, who were evaluated for HIV drug resistance (HIVDR) and other risk factors. Setting In 2018-2020, pregnant (weeks-14-28) women living with HIV and ≤14 days of ART were enrolled at 22 international sites and followed with their infants through 50 weeks postpartum. Methods HIV sequences derived by single genome amplification (SGA) from longitudinally collected specimens were assessed from VT Cases for HIVDR in protease, reverse transcriptase, integrase, and the nef 3'polypurine tract (3'PPT). Results The four Case mothers were prescribed efavirenz-based-ART for 1-7 days prior to randomization to study ART. Their infants received postnatal nevirapine+/-zidovudine prophylaxis and were breastfed. A total of 833 SGA sequences were derived. The "major" (Stanford HIVDR Score ≥60) non-nucleoside reverse transcriptase inhibitor (NNRTI) mutation (K103N) was detected persistently in one viremic mother, and likely contributed to VT of HIVDR. Major NNRTI HIVDR mutations were detected in all three surviving infants. No integrase, nor high frequencies of 3'PPT mutations conferring dolutegravir HIVDR were detected. The timing of HIV infant diagnosis, plasma HIV RNA levels and HIVDR suggests one in utero, one peripartum, one early, and one late breastfeeding transmission. Conclusions VT was rare. New-onset NNRTI HIVDR in Case mothers was likely from efavirenz-ART prescribed prior to study dolutegravir-ART, and in one case appeared transmitted to the infant despite nevirapine prophylaxis.
Collapse
Affiliation(s)
- Marley D. Bishop
- Department of Global Infectious Diseases, Seattle Children’s Research Institute, 307 Westlake Ave N, Seattle, 98109, Washington, USA
| | - Violet Korutaro
- Children’s Foundation Uganda, Baylor College of Medicine, Block 5 Mulago Hospital, P. O. BOX 72052, Kampala 72052, Kamutarpala Uganda
| | - Ceejay L. Boyce
- Department of Global Infectious Diseases, Seattle Children’s Research Institute, 307 Westlake Ave N, Seattle, 98109, Washington, USA
| | - Ingrid A. Beck
- Department of Global Infectious Diseases, Seattle Children’s Research Institute, 307 Westlake Ave N, Seattle, 98109, Washington, USA
| | - Sheila M. Styrchak
- Department of Global Infectious Diseases, Seattle Children’s Research Institute, 307 Westlake Ave N, Seattle, 98109, Washington, USA
| | - Kevin Knowles
- Frontier Science and Technology Research Foundation, 4033 Maple Road Amherst, Buffalo, 14226, NY, USA
| | - Lauren Ziemba
- Centre for Biostatistics in AIDS Research Center for Biostatistics in AIDS Research, Harvard University T.H. Chan School of Public Health, FXB 507 677 Huntington Ave Center for Biostatistics in AIDS Research, Boston, 02115, MA, USA
| | - Sean S. Brummel
- Centre for Biostatistics in AIDS Research Center for Biostatistics in AIDS Research, Harvard University T.H. Chan School of Public Health, FXB 507 677 Huntington Ave Center for Biostatistics in AIDS Research, Boston, 02115, MA, USA
| | - Anne Coletti
- FHI 360, 359 Blackwell St. Suite 200, Durham, 27713, NC, USA
| | - Patrick Jean-Philippe
- Division of AIDS; Maternal Adolescent Pediatric Research Branch; Prevention Sciences Program, National Institute of Allergy and Infectious Diseases, 5601 Fishers Lane Room 8B21, MSC 9831, Bethesda, 20892, MD, USA
| | - Nahida Chakhtoura
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Maternal and Pediatric Infectious Disease Branch, 6710B Rockledge Drive, Bethesda, MD, USA 20892
| | - Tichaona Vhembo
- University of Zimbabwe-University of California San Francisco Collaborative Research Program (UZ-UCSF), 15 Phillips Ave, Belgravia Harare, Zimbabwe
| | - Haseena Cassim
- Perinatal HIV Research Unit, University of the Witwatersrand Johannesburg, Chris Hani Baragwanath Hospital P.O. Box 114, Diepkloof, 1864, Johannesburg, 2050, Gauteng, South Africa
| | - Maxensia Owor
- Makerere University –John Hopkins University Research Collaboration (MUJHU CARE LTD), CRS Upper Mulago Hill Road PO Box 23491, Kampala, Uganda
| | - Lee Fairlie
- Wits RHI, Maternal and Child Health, 22 Esselen Street Hillbrow, Johannesburg, Gauteng, South Africa 2001
| | - Sikhulile Moyo
- Botswana-Harvard AIDS Institute Partnership, Plot 1836 N Ring Rd, Gaborone, Botswana
- Division of Infectious Disease, Brigham and Women’s Hospital, 15 Francis St 2nd Floor, Boston, 02115, MA, USA
| | - Lameck Chinula
- Division of Global Women’s Health; Department of Obstetrics and Gynecology, University of North Carolina-Chapel Hill, 3009 Old Clinic Building Campus Box 7570, Chapel Hill, 27599, NC, USA
| | - Shahin Lockman
- Botswana-Harvard AIDS Institute Partnership, Plot 1836 N Ring Rd, Gaborone, Botswana
- Division of Infectious Disease, Brigham and Women’s Hospital, 15 Francis St 2nd Floor, Boston, 02115, MA, USA
- Harvard University T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases School of Public Health, 655 Huntington Ave, Boston, 02115, MA, USA
| | - Lisa M. Frenkel
- Department of Global Infectious Diseases, Seattle Children’s Research Institute, 307 Westlake Ave N, Seattle, 98109, Washington, USA
- University of Washington, Department of Global Health, Medicine, Epidemiology and Pediatrics, 1959 NE Pacific St. Seattle 98195 WA
| |
Collapse
|
9
|
Devendra A, Kohler M, Letsika M, Khooa H, Motaboli L, Lerotholi M, Tschumi N, Labhardt ND, Brown JA. HIV viral suppression in children and adolescents 2 years after transition to dolutegravir: a multicentre cohort study. AIDS 2024; 38:1013-1023. [PMID: 38381717 PMCID: PMC11064912 DOI: 10.1097/qad.0000000000003835] [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: 09/20/2023] [Revised: 12/23/2023] [Accepted: 01/04/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Treatment failure is common among children and adolescents with HIV. Antiretroviral therapy (ART) containing dolutegravir has recently been rolled out across Africa, though long-term real-world data in paediatric populations are lacking. Here, we report treatment outcomes among children and adolescents in Lesotho who transitioned from nonnucleoside reverse transcriptase inhibitor (NNRTI)-based to dolutegravir-based ART through 2 years' follow-up. METHODS Data were derived from two open cohort studies in Lesotho. Children and adolescents aged less than 18 years who transitioned from NNRTI-based to dolutegravir-based ART at least 18 months before data closure were included. We report viral load results less than 12 months before, 12 (window: 6-17) months after, and 24 (window: 18-29) months after transition to dolutegravir. Associations of pretransition demographic and clinical factors with 24-month viraemia were assessed through multivariable logistic regression. RESULTS Among 2126 included individuals, 1100 (51.7%) were female individuals, median age at transition to dolutegravir was 14.0 years [interquartile range (IQR) 11.5-15.8], and median time taking ART at transition was 7.6 years (IQR 4.4-10.6). Among those with a viral load result at the respective time points, viral suppression to less than 50 copies/ml was achieved by 1635 of 1973 (82.9%) less than 12 months before, 1846 of 2012 (91.8%) 12 months after, and 1725 of 1904 (90.6%) 24 months after transition to dolutegravir. Pretransition viraemia was associated with viraemia at 24 months, though more than 80% of individuals with pretransition viraemia achieved resuppression to less than 50 copies/ml at 24 months. CONCLUSION The proportion of children and adolescents with viral suppression increased after transition to dolutegravir, though further progress is needed to reach global targets.
Collapse
Affiliation(s)
- Akash Devendra
- Department of Pediatrics, Baylor College of Medicine
- Baylor International Pediatric AIDS Initiative, Houston, TX, USA
- Baylor College of Medicine Children's Foundation Lesotho, Maseru, Lesotho
| | - Maurus Kohler
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel
- University of Basel, Basel, Switzerland
| | - Motlatsi Letsika
- Baylor College of Medicine Children's Foundation Lesotho, Maseru, Lesotho
| | - Hape Khooa
- Baylor College of Medicine Children's Foundation Lesotho, Maseru, Lesotho
| | | | - Malebanye Lerotholi
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel
- University of Basel, Basel, Switzerland
- Ministry of Health of Lesotho, Maseru, Lesotho
| | - Nadine Tschumi
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel
- University of Basel, Basel, Switzerland
| | - Niklaus D. Labhardt
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel
- University of Basel, Basel, Switzerland
| | - Jennifer A. Brown
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital Basel
- University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Nakatudde I, Katana E, Agnes Odongpiny EL, Nalugga EA, Castelnuovo B, Fowler MG, Musoke P. Prevalence of overweight and obesity among adolescents living with HIV after dolutegravir - based antiretroviral therapy start in Kampala, Uganda. AIDS Res Ther 2024; 21:23. [PMID: 38637785 PMCID: PMC11027216 DOI: 10.1186/s12981-024-00615-6] [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: 07/26/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Dolutegravir (DTG)-based antiretroviral therapy (ART) is currently the preferred first-line treatment for persons living with HIV (PLHIV) including children and adolescents in many low- and middle-income countries including Uganda. However, there are concerns about excessive weight gain associated with DTG especially in adults. There remains paucity of current information on weight-related outcomes among adolescents on DTG. We determined the prevalence of excessive weight gain and associated factors among adolescents living with HIV (ALHIV) receiving DTG-based ART in Kampala, Uganda. METHODS Cross-sectional study involving ALHIV aged 10-19 years on DTG-based ART for at least one year were recruited from public health facilities in Kampala between February and May 2022. Excessive weight gain was defined as becoming overweight or obese per body mass index (BMI) norms while on DTG-based ART for at least one year. Demographic, clinical and laboratory data were collected using interviewer-administered questionnaires and data extracted from medical records. At enrolment, blood pressure and anthropometry were measured and blood was drawn for blood glucose and lipid profile. Data was summarised using descriptive statistics and logistic regression was performed to determine the associated factors. RESULTS We enrolled 165 ALHIV with a median age of 14 years (IQR 12-16). Eighty (48.5%) were female. The median duration on ART and DTG was 8 years (IQR 7-11) and 2 years (IQR 1-3) respectively. At DTG initiation, the majority of participants (152/165, 92.1%) were ART-experienced, and had normal BMI (160/165, 97%). Overall, 12/165 (7.3%) adolescents (95% CI: 4.2-12.4) had excessive weight gain. No factors were significantly associated with excessive weight gain after DTG start in ALHIV. However, all ALHIV with excessive weight gain were females. CONCLUSION Our study found a prevalence of 7.3% of overweight and obesity in ALHIV after initiating DTG. We did not find any factor significantly associated with excessive weight gain in ALHIV on DTG. Nonetheless, we recommend ongoing routine monitoring of anthropometry and metabolic markers in ALHIV as DTG use increases globally, to determine the exact magnitude of excessive weight gain and to identify those at risk of becoming overweight or obese while taking the medication.
Collapse
Affiliation(s)
- Irene Nakatudde
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda.
| | - Elizabeth Katana
- College of Health Sciences, Makerere University, Kampala, Uganda
| | - Eva Laker Agnes Odongpiny
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
- School of Medicine, University of St. Andrews, St. Andrews, Scotland
| | - Esther Alice Nalugga
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Barbara Castelnuovo
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Mary Glenn Fowler
- Department of Pathology, School of Medicine, John Hopkins University, Baltimore, MD, USA
| | - Philippa Musoke
- Makerere University-John Hopkins University Research Collaboration, Kampala, Uganda
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
11
|
Spector SA, Brummel SS, Chang A, Wiznia A, Ruel TD, Acosta EP, IMPAACT P1093 Team. Impact of Genetic Variants in ABCG2 , NR1I2 , and UGT1A1 on the Pharmacokinetics of Dolutegravir in Children. J Acquir Immune Defic Syndr 2024; 95:297-303. [PMID: 38180896 PMCID: PMC10922521 DOI: 10.1097/qai.0000000000003358] [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: 03/06/2023] [Accepted: 08/22/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Dolutegravir plasma concentrations and pharmacokinetic (PK) parameters in children display considerable variability. Here, the impact of genetic variants in ABCG2 421C>A (rs2231142), NR1I2 63396 C>T (rs2472677), and UGT1A1 (rs5839491) on dolutegravir PK was examined. METHODS Children defined by age and administered dolutegravir formulation had AUC 24 at steady state, C max and C 24h determined. Associations between genetic variants and PK parameters were assessed using the dominant inheritance model. RESULTS The 59 children studied had a median age of 4.6 years, log 10 plasma HIV RNA of 4.79 (copies/mm 3 ), and CD4 + lymphocyte count of 1041 cells/mm 3 ; 51% were female. For ABCG2 , participants with ≥1 minor allele had lower adjusted mean AUC difference (hr*mg/L) controlling for weight at entry, cohort and sex (-15.7, 95% CI: [-32.0 to 0.6], P = 0.06), and log 10 C max adjusted mean difference (-0.15, 95% CI: [-0.25 to -0.05], P = 0.003). Participants with ≥1 minor allele had higher adjusted mean AUC difference (11.9, 95% CI: [-1.1 to 25.0], P = 0.07). For UGT1A1 , poor metabolizers had nonsignificant higher concentrations (adjusted log 10 C max mean difference 11.8; 95% CI: [-12.3 to 36.0], P = 0.34) and lower mean log 10 adjusted oral clearance -0.13 L/h (95% CI: [-0.3 to 0.06], P = 0.16). No association was identified between time-averaged AUC differences by genotype for adverse events, plasma HIV RNA, or CD4 + cell counts. CONCLUSIONS Dolutegravir AUC 24 for genetic variants in ABCG2 , NR1l2 , and UGT1A1 varied from -25% to +33%. These findings help to explain some of the variable pharmacokinetics identified with dolutegravir in children.
Collapse
Affiliation(s)
- Stephen A. Spector
- University of California San Diego, La Jolla, CA and Rady Children’s Hospital San Diego, San Diego, CA
| | - Sean S. Brummel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health
| | - Audrey Chang
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health
| | - Andrew Wiznia
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | | | | | | |
Collapse
|
12
|
Chandiwana NC, Siedner MJ, Marconi VC, Hill A, Ali MK, Batterham RL, Venter WDF. Weight Gain After HIV Therapy Initiation: Pathophysiology and Implications. J Clin Endocrinol Metab 2024; 109:e478-e487. [PMID: 37437159 PMCID: PMC10795932 DOI: 10.1210/clinem/dgad411] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 06/14/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023]
Abstract
Rapid advances in the potency, safety, and availability of modern HIV antiretroviral therapy (ART) have yielded a near-normal life expectancy for most people living with HIV (PLWH). Ironically, considering the history of HIV/AIDS (initially called "slim disease" because of associated weight loss), the latest dilemma faced by many people starting HIV therapy is weight gain and obesity, particularly Black people, women, and those who commenced treatment with advanced immunodeficiency. We review the pathophysiology and implications of weight gain among PLWH on ART and discuss why this phenomenon was recognized only recently, despite the availability of effective therapy for nearly 30 years. We comprehensively explore the theories of the causes, from initial speculation that weight gain was simply a return to health for people recovering from wasting to comparative effects of newer regimens vs prior toxic agents, to direct effects of agents on mitochondrial function. We then discuss the implications of weight gain on modern ART, particularly concomitant effects on lipids, glucose metabolism, and inflammatory markers. Finally, we discuss intervention options for PLWH and obesity, from the limitations of switching ART regimens or specific agents within regimens, weight-gain mitigation strategies, and potential hope in access to emerging antiobesity agents, which are yet to be evaluated in this population.
Collapse
Affiliation(s)
- Nomathemba C Chandiwana
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Mark J Siedner
- Medical Practice Evaluation Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Vincent C Marconi
- Division of Infectious Diseases and Department of Global Health, Emory University School of Medicine and Rollins School of Public Health, Atlanta, GA 4223, USA
| | - Andrew Hill
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 7BE, UK
| | - Mohammed K Ali
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 4223, USA
- Department of Family and Preventive Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | | | - Willem Daniel Francois Venter
- Ezintsha, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
- Department of Public Health Medicine, Faculty of Health Sciences, School of Health Systems and Public Health, University of Pretoria, Pretoria 0028, South Africa
| |
Collapse
|
13
|
De Clercq E, Zhang Z, Huang J, Zhang M, Li G. Biktarvy for the treatment of HIV infection: Progress and prospects. Biochem Pharmacol 2023; 217:115862. [PMID: 37858869 DOI: 10.1016/j.bcp.2023.115862] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Bictegravir (BIC), a second-generation integrase strand-transfer inhibitor (INSTI) with high resilience to INSTI-resistance mutations, is integrated as a key component of Biktarvy® - a fixed-dose once-daily triple-drug regimen of bictegravir (BIC), emtricitabine (FTC) plus tenofovir alafenamide (TAF). Based on the accumulated evidence from HIV clinical trials and real-world studies, the clinical effectiveness of BIC + FTC + TAF has been proven non-inferior to other fixed-dose once-daily combinations such as dolutegravir + FTC + TAF and dolutegravir + abacavir + lamivudine. Biktarvy also shows limited drug-drug interactions and a high barrier to drug resistance. According to recent HIV guidelines, BIC + FTC + TAF is recommended as initial and long-term therapy for the treatment of HIV infection. For the pre-exposure prophylaxis, tenofovir disoproxil fumarate (TDF) or tenofovir alafenamide (TAF) remains advisable, but BIC may be possibly added to TDF or TAF. In the development of a long-acting once-monthly regimen, the novel nano-formulation of BIC + FTC + TAF could be possibly developed in the future.
Collapse
Affiliation(s)
- Erik De Clercq
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven 3000, Belgium
| | - Zhenlan Zhang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Jie Huang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410013, China
| | - Min Zhang
- Institute of Hepatology and Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Guangdi Li
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410013, China; FuRong Laboratory, Changsha 410078, China.
| |
Collapse
|
14
|
Gill MM, Herrera N, Guilaze R, Mussa A, Dengo N, Nhangave A, Mussá J, Perez P, Bhatt N. Virologic Outcomes and ARV Switch Profiles 2 Years After National Rollout of Dolutegravir to Children Less Than 15 Years in Southern Mozambique. Pediatr Infect Dis J 2023; 42:893-898. [PMID: 37409808 DOI: 10.1097/inf.0000000000004037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
BACKGROUND Dolutegravir (DTG) was scaled up globally to optimize treatment for children living with HIV. We evaluated the rollout and virological outcomes after DTG introduction in Mozambique. METHODS Data from children 0-14 years with visits from September 2019 to August 2021 were extracted from records in 16 facilities in 12 districts. Among children ever on DTG, we report treatment switches, defined as changes in anchor drug, regardless of changes to nucleoside reverse transcriptase inhibitor (NRTI) backbones. Among those on DTG for ≥6 months, we described viral load suppression rates by children newly initiating and switching to DTG and by the NRTI backbone at the time of the DTG switch. RESULTS Overall, 3,347 children were ever on DTG-based treatment (median age 9.5 years; 52.8% female). Most children (3,202, 95.7%) switched to DTG from another antiretroviral regimen. During the 2-year follow-up, 9.9% never switched from DTG; 52.7% had 1 regimen change, of which 97.6% were switched to DTG. However, 37.2% of children experienced ≥2 anchor drug changes. Overall median time on DTG was 18.6 months; nearly all children ≥5 years (98.6%) were on DTG at the last visit. Viral suppression was 79.7% (63/79) for children newly initiating DTG and 85.8% (1,775/2,068) for those switching to DTG. Suppression rates were 84.8% and 85.7% among children who switched and maintained NRTI backbones, respectively. CONCLUSIONS Viral suppression rates of ≥80% with minor variations by backbone were achieved during the 2-year DTG rollout. However, there were multiple anchor drug switches for over one-third of children, which may be attributable in part to drug stockouts. Long-term pediatric HIV management will only be successful with immediate and sustainable access to optimized child-friendly drugs and formulations.
Collapse
Affiliation(s)
- Michelle M Gill
- From the Research Department, Elizabeth Glaser Pediatric AIDS Foundation, Washington DC
| | - Nicole Herrera
- From the Research Department, Elizabeth Glaser Pediatric AIDS Foundation, Washington DC
| | - Rui Guilaze
- Research Department, Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Abdul Mussa
- Research Department, Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Nataniel Dengo
- Research Department, Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Amancio Nhangave
- Gaza Provincial Health Directorate, Mozambique Ministry of Health
| | - Jaciara Mussá
- Inhambane Provincial Health Directorate, Mozambique Ministry of Health
| | - Patricia Perez
- Research Department, Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| | - Nilesh Bhatt
- Research Department, Elizabeth Glaser Pediatric AIDS Foundation, Maputo, Mozambique
| |
Collapse
|