Pub Date : 2025-01-01DOI: 10.1016/S2352-3018(24)00348-5
The Lancet Hiv
{"title":"Bleak prospects for HIV response under new US administration.","authors":"The Lancet Hiv","doi":"10.1016/S2352-3018(24)00348-5","DOIUrl":"https://doi.org/10.1016/S2352-3018(24)00348-5","url":null,"abstract":"","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":"12 1","pages":"e1"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142957283","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-06-17DOI: 10.1016/S2352-3018(24)00159-0
Talha Burki
{"title":"Recent gay Black history in the UK.","authors":"Talha Burki","doi":"10.1016/S2352-3018(24)00159-0","DOIUrl":"10.1016/S2352-3018(24)00159-0","url":null,"abstract":"","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":"e12"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141433160","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-12-10DOI: 10.1016/S2352-3018(24)00271-6
Mansoor Farahani, Shannon M Farley, Theodore F Smart, Felix Ndagije, Limpho Maile, Herbert Longwe, David Hoos, Wafaa M El-Sadr
Background: Lesotho has made substantial efforts to control its HIV epidemic. We aimed to assess progress towards UNAIDS 95-95-95 targets in Lesotho by comparing data from the Lesotho Population-based HIV Impact Assessments conducted in 2016-17 (LePHIA 2016) and 2019-20 (LePHIA 2020).
Methods: The LePHIA surveys used a cross-sectional, two-stage, stratified cluster sampling design to obtain a nationally representative sample of adults aged 15-59 years (LePHIA 2016) or aged 15 years and older (LePHIA 2020) from all ten districts of Lesotho. From November, 2016 to May, 2017 (LePHIA 2016) and from December, 2019 to March, 2020 (LePHIA 2020), consenting participants from randomly selected households provided demographic and clinical information and blood samples for household HIV testing according to national guidelines. HIV-reactive test results were confirmed by a laboratory assay. We estimated HIV status awareness and antiretroviral therapy (ART) use on the basis of self-reports or detection of antiretroviral drugs in blood samples. Viral load suppression was defined as HIV-1 RNA less than 1000 copies per mL. We applied Poisson regression models using survey weights and estimated variances using the Taylor series linearisation approach.
Findings: 11 682 participants were enrolled in LePHIA 2016 and 12 718 participants were enrolled in LePHIA 2020. Overall HIV incidence decreased significantly from 1·10% (95% CI 0·68-1·52) in 2016 to 0·50% (0·26-0·74) in 2020 (p=0·026). Among adults who tested positive for HIV, awareness of HIV status improved from 81·0% (79·6-82·3) in 2016 to 89·6% (88·3-90·8) in 2020 (p<0·0001). Furthermore, between the two surveys, the proportion on ART among those aware of their HIV status increased from 91·8% (90·5-93·0) to 96·9% (95·9-97·6; p<0·0001) and the prevalence of viral load suppression among those on ART increased from 87·7% (86·1-89·1) to 90·8% (89·5-91·9; p<0·0020). After adjusting for demographic covariates, adults living with HIV were significantly more likely in 2020 than in 2016 to know their HIV status (adjusted prevalence ratio 1·09, 95% CI 1·07-1·12, p<0·0001), to be on ART if aware of their status (1·05, 1·03-1·07, p<0·0001), and to be virally suppressed if on ART (1·03, 1·01-1·06, p=0·0045).
Interpretation: Between 2016 and 2020, Lesotho made significant progress towards the UNAIDS 95-95-95 targets, surpassing the second target (ART coverage) and showing improvements in HIV status awareness and viral load suppression (the first and third targets) as well as declines in HIV prevalence and incidence. Lesotho's experience provides valuable insights for other countries working to control their HIV epidemics.
Funding: The US President's Emergency Plan for AIDS Relief.
{"title":"Lesotho's progress towards UNAIDS 95-95-95 targets from 2016 to 2020: comparison of Population-based HIV Impact Assessments.","authors":"Mansoor Farahani, Shannon M Farley, Theodore F Smart, Felix Ndagije, Limpho Maile, Herbert Longwe, David Hoos, Wafaa M El-Sadr","doi":"10.1016/S2352-3018(24)00271-6","DOIUrl":"10.1016/S2352-3018(24)00271-6","url":null,"abstract":"<p><strong>Background: </strong>Lesotho has made substantial efforts to control its HIV epidemic. We aimed to assess progress towards UNAIDS 95-95-95 targets in Lesotho by comparing data from the Lesotho Population-based HIV Impact Assessments conducted in 2016-17 (LePHIA 2016) and 2019-20 (LePHIA 2020).</p><p><strong>Methods: </strong>The LePHIA surveys used a cross-sectional, two-stage, stratified cluster sampling design to obtain a nationally representative sample of adults aged 15-59 years (LePHIA 2016) or aged 15 years and older (LePHIA 2020) from all ten districts of Lesotho. From November, 2016 to May, 2017 (LePHIA 2016) and from December, 2019 to March, 2020 (LePHIA 2020), consenting participants from randomly selected households provided demographic and clinical information and blood samples for household HIV testing according to national guidelines. HIV-reactive test results were confirmed by a laboratory assay. We estimated HIV status awareness and antiretroviral therapy (ART) use on the basis of self-reports or detection of antiretroviral drugs in blood samples. Viral load suppression was defined as HIV-1 RNA less than 1000 copies per mL. We applied Poisson regression models using survey weights and estimated variances using the Taylor series linearisation approach.</p><p><strong>Findings: </strong>11 682 participants were enrolled in LePHIA 2016 and 12 718 participants were enrolled in LePHIA 2020. Overall HIV incidence decreased significantly from 1·10% (95% CI 0·68-1·52) in 2016 to 0·50% (0·26-0·74) in 2020 (p=0·026). Among adults who tested positive for HIV, awareness of HIV status improved from 81·0% (79·6-82·3) in 2016 to 89·6% (88·3-90·8) in 2020 (p<0·0001). Furthermore, between the two surveys, the proportion on ART among those aware of their HIV status increased from 91·8% (90·5-93·0) to 96·9% (95·9-97·6; p<0·0001) and the prevalence of viral load suppression among those on ART increased from 87·7% (86·1-89·1) to 90·8% (89·5-91·9; p<0·0020). After adjusting for demographic covariates, adults living with HIV were significantly more likely in 2020 than in 2016 to know their HIV status (adjusted prevalence ratio 1·09, 95% CI 1·07-1·12, p<0·0001), to be on ART if aware of their status (1·05, 1·03-1·07, p<0·0001), and to be virally suppressed if on ART (1·03, 1·01-1·06, p=0·0045).</p><p><strong>Interpretation: </strong>Between 2016 and 2020, Lesotho made significant progress towards the UNAIDS 95-95-95 targets, surpassing the second target (ART coverage) and showing improvements in HIV status awareness and viral load suppression (the first and third targets) as well as declines in HIV prevalence and incidence. Lesotho's experience provides valuable insights for other countries working to control their HIV epidemics.</p><p><strong>Funding: </strong>The US President's Emergency Plan for AIDS Relief.</p>","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":"e51-e59"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142822745","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-11-29DOI: 10.1016/S2352-3018(24)00248-0
Lucette A Cysique, Jules Levin, Chris Howard, Jeff Taylor, John Rule, Jane Costello, Jane Bruning, Priscilla Njeri, Amy B Mullens, Edwina Wright, Hetta Gouse, Kirstie Daken, Mattia Trunfio, Htein Linn Aung, Reuben N Robbins, Christopher M Ferraris, Jose A Muñoz-Moreno, Steven P Woods, David J Moore, Christopher Power, Pui Li Wong, Kejal Hasmukharay, Primrose Nyamayaro, Jaime Vera, Reena Rajasuriar, Robert K Heaton, Karl Goodkin, Scott Letendre, Ronald J Ellis, Bruce J Brew, Sean B Rourke
Prevalence and incidence of HIV among people aged 50 years and older continue to rise worldwide, generating increasing awareness among care providers, scientists, and the HIV community about the importance of brain health in older adults with HIV. Many age-related factors that adversely affect brain health can occur earlier and more often among people with HIV, including epigenetic ageing, chronic medical conditions (eg, cardiovascular disease), and age-related syndromes (eg, frailty). Extensive dialogue between HIV community leaders, health-care providers, and scientists has led to the development of a multidimensional response strategy to protect and enhance brain health in people ageing with HIV that spans across public health, clinical spaces, and research spaces. This response strategy was informed by integrated ageing care frameworks and is centred on prevention, early detection, and management of brain health issues associated with HIV (eg, neurocognitive disorders), with specific considerations for low-resource or middle-resource countries. A collaborative, international, and data-informed update of the diagnostic criteria for HIV-associated neurocognitive disorders is a cornerstone of the proposed response strategy. The proposed response strategy includes a dynamic, international, online knowledge hub that will provide a crucial community resource for emerging evidence on the brain health of people ageing with HIV.
{"title":"Fostering healthy cognitive ageing in people living with HIV.","authors":"Lucette A Cysique, Jules Levin, Chris Howard, Jeff Taylor, John Rule, Jane Costello, Jane Bruning, Priscilla Njeri, Amy B Mullens, Edwina Wright, Hetta Gouse, Kirstie Daken, Mattia Trunfio, Htein Linn Aung, Reuben N Robbins, Christopher M Ferraris, Jose A Muñoz-Moreno, Steven P Woods, David J Moore, Christopher Power, Pui Li Wong, Kejal Hasmukharay, Primrose Nyamayaro, Jaime Vera, Reena Rajasuriar, Robert K Heaton, Karl Goodkin, Scott Letendre, Ronald J Ellis, Bruce J Brew, Sean B Rourke","doi":"10.1016/S2352-3018(24)00248-0","DOIUrl":"10.1016/S2352-3018(24)00248-0","url":null,"abstract":"<p><p>Prevalence and incidence of HIV among people aged 50 years and older continue to rise worldwide, generating increasing awareness among care providers, scientists, and the HIV community about the importance of brain health in older adults with HIV. Many age-related factors that adversely affect brain health can occur earlier and more often among people with HIV, including epigenetic ageing, chronic medical conditions (eg, cardiovascular disease), and age-related syndromes (eg, frailty). Extensive dialogue between HIV community leaders, health-care providers, and scientists has led to the development of a multidimensional response strategy to protect and enhance brain health in people ageing with HIV that spans across public health, clinical spaces, and research spaces. This response strategy was informed by integrated ageing care frameworks and is centred on prevention, early detection, and management of brain health issues associated with HIV (eg, neurocognitive disorders), with specific considerations for low-resource or middle-resource countries. A collaborative, international, and data-informed update of the diagnostic criteria for HIV-associated neurocognitive disorders is a cornerstone of the proposed response strategy. The proposed response strategy includes a dynamic, international, online knowledge hub that will provide a crucial community resource for emerging evidence on the brain health of people ageing with HIV.</p>","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":"e71-e80"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142773979","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-12-05DOI: 10.1016/S2352-3018(24)00264-9
Kombatende Sikombe, Aaloke Mody, Charles W Goss, Sandra Simbeza, Laura K Beres, Jake M Pry, Ingrid Eshun-Wilson, Anjali Sharma, Njekwa Mukamba, Lloyd B Mulenga, Brian Rice, Jacob Mutale, Alida Zulu Dube, Musunge Mulabe, James Hargreaves, Carolyn Bolton Moore, Charles B Holmes, Izukanji Sikazwe, Elvin H Geng
<p><strong>Background: </strong>Recipients of health services value not only convenience but also respectful, kind, and helpful providers. To date, research to improve person-centred HIV treatment has focused on making services easier to access (eg, differentiated service delivery) rather than the interpersonal experience of care. We developed and evaluated a person-centred care (PCC) intervention targeting practices of health-care workers.</p><p><strong>Methods: </strong>Using a stepped-wedge, cluster-randomised design, we randomly allocated 24 HIV clinics stratified by size in Zambia into four groups and introduced a PCC intervention that targeted caring aspects of the behaviour of health-care workers in one group every 6 months. The intervention entailed training and coaching for health-care workers on PCC practices (to capacitate), client experience assessment with feedback to facilities (to create opportunities), and small performance-based incentives (to motivate). In a probability sample of clients who were pre-trained on a client experience exit survey and masked to facility intervention status, we evaluated effects on client experience by use of mean score change and also proportion with poor encounters (ie, score of ≤8 on a 12-point survey instrument). We examined effects on missed visits (ie, >30 days late for next scheduled encounter) in all groups and retention in care at 15 months in group 1 and group 4 by use of electronic health records. We assessed effects on treatment success at 15 months (ie, HIV RNA concentration <400 copies per mL or adjudicated care status) in a prospectively enrolled subset of clients from group 1 and group 4. We estimated treatment effects with mixed-effects logistic regression, adjusting for sex, age, and baseline care status. This trial is registered at the Pan-African Clinical Trials Registry (202101847907585), and is completed.</p><p><strong>Findings: </strong>Between Aug 12, 2019, and Nov 30, 2021, 177 543 unique clients living with HIV made at least one visit to one of the 24 study clinics. The PCC intervention reduced the proportion of poor visits based on exit surveys from 147 (23·3%) of 632 during control periods to 33 (13·3%) of 249 during the first 6 months of intervention, and then to eight (3·5%) of 230 at 6 months or later (adjusted risk difference [aRD] for control vs ≥6 months intervention -16·9 percentage points, 95% CI -24·8 to -8·9). Among all adult scheduled appointments, the PCC intervention reduced the proportion of missed visits from 90 593 (25·3%) of 358 741 during control periods to 40 380 (22·6%) of 178 523 in the first 6 months, and then 52 288 (21·5%) of 243 350 at 6 months or later (aRD for control vs the intervention -4·2 percentage points, 95% CI -4·8 to -3·7). 15-month retention improved from 33 668 (80·2%) of 41 998 in control to 35 959 (83·6%) of 43 005 during intervention (aRD 5·9 percentage points, 95% CI 0·6 to 11·2), with larger effects in clients newly starting treatm
{"title":"Effect of a multicomponent, person-centred care intervention on client experience and HIV treatment outcomes in Zambia: a stepped-wedge, cluster-randomised trial.","authors":"Kombatende Sikombe, Aaloke Mody, Charles W Goss, Sandra Simbeza, Laura K Beres, Jake M Pry, Ingrid Eshun-Wilson, Anjali Sharma, Njekwa Mukamba, Lloyd B Mulenga, Brian Rice, Jacob Mutale, Alida Zulu Dube, Musunge Mulabe, James Hargreaves, Carolyn Bolton Moore, Charles B Holmes, Izukanji Sikazwe, Elvin H Geng","doi":"10.1016/S2352-3018(24)00264-9","DOIUrl":"10.1016/S2352-3018(24)00264-9","url":null,"abstract":"<p><strong>Background: </strong>Recipients of health services value not only convenience but also respectful, kind, and helpful providers. To date, research to improve person-centred HIV treatment has focused on making services easier to access (eg, differentiated service delivery) rather than the interpersonal experience of care. We developed and evaluated a person-centred care (PCC) intervention targeting practices of health-care workers.</p><p><strong>Methods: </strong>Using a stepped-wedge, cluster-randomised design, we randomly allocated 24 HIV clinics stratified by size in Zambia into four groups and introduced a PCC intervention that targeted caring aspects of the behaviour of health-care workers in one group every 6 months. The intervention entailed training and coaching for health-care workers on PCC practices (to capacitate), client experience assessment with feedback to facilities (to create opportunities), and small performance-based incentives (to motivate). In a probability sample of clients who were pre-trained on a client experience exit survey and masked to facility intervention status, we evaluated effects on client experience by use of mean score change and also proportion with poor encounters (ie, score of ≤8 on a 12-point survey instrument). We examined effects on missed visits (ie, >30 days late for next scheduled encounter) in all groups and retention in care at 15 months in group 1 and group 4 by use of electronic health records. We assessed effects on treatment success at 15 months (ie, HIV RNA concentration <400 copies per mL or adjudicated care status) in a prospectively enrolled subset of clients from group 1 and group 4. We estimated treatment effects with mixed-effects logistic regression, adjusting for sex, age, and baseline care status. This trial is registered at the Pan-African Clinical Trials Registry (202101847907585), and is completed.</p><p><strong>Findings: </strong>Between Aug 12, 2019, and Nov 30, 2021, 177 543 unique clients living with HIV made at least one visit to one of the 24 study clinics. The PCC intervention reduced the proportion of poor visits based on exit surveys from 147 (23·3%) of 632 during control periods to 33 (13·3%) of 249 during the first 6 months of intervention, and then to eight (3·5%) of 230 at 6 months or later (adjusted risk difference [aRD] for control vs ≥6 months intervention -16·9 percentage points, 95% CI -24·8 to -8·9). Among all adult scheduled appointments, the PCC intervention reduced the proportion of missed visits from 90 593 (25·3%) of 358 741 during control periods to 40 380 (22·6%) of 178 523 in the first 6 months, and then 52 288 (21·5%) of 243 350 at 6 months or later (aRD for control vs the intervention -4·2 percentage points, 95% CI -4·8 to -3·7). 15-month retention improved from 33 668 (80·2%) of 41 998 in control to 35 959 (83·6%) of 43 005 during intervention (aRD 5·9 percentage points, 95% CI 0·6 to 11·2), with larger effects in clients newly starting treatm","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":"e26-e39"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142796571","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-10-29DOI: 10.1016/S2352-3018(24)00270-4
Thibaut Vanbaelen, Achilleas Tsoumanis, Chris Kenyon
{"title":"Screening for chlamydia and incidence of symptomatic infections.","authors":"Thibaut Vanbaelen, Achilleas Tsoumanis, Chris Kenyon","doi":"10.1016/S2352-3018(24)00270-4","DOIUrl":"10.1016/S2352-3018(24)00270-4","url":null,"abstract":"","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":"e9"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142564961","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-12-10DOI: 10.1016/S2352-3018(24)00247-9
Srilatha Edupuganti, Christopher B Hurt, Kathryn E Stephenson, Yunda Huang, Carmen A Paez, Chenchen Yu, Catherine Yen, Brett Hanscom, Zonglin He, Maurine D Miner, Theresa Gamble, Jack Heptinstall, Kelly E Seaton, Elizabeth Domin, Bob C Lin, Krisha McKee, Nicole Doria-Rose, Stephanie Regenold, Hans Spiegel, Maija Anderson, Nadia McClosky, Lily Zhang, Estelle Piwowar-Manning, Margaret E Ackerman, Michael Pensiero, Bonnie J Dye, Raphael J Landovitz, Kenneth Mayer, Marc Siegel, Magdalena Sobieszczyk, Stephen R Walsh, Lucio Gama, Dan H Barouch, David C Montefiori, Georgia D Tomaras
<p><strong>Background: </strong>Multiple broadly neutralising monoclonal antibodies (mAbs) are in development for HIV-1 prevention. The aim of this trial was to test the PGT121.414.LS and VRC07-523LS mAbs for safety and pharmacokinetics in adults.</p><p><strong>Methods: </strong>In this first-in-human phase 1 trial (HVTN 136/HPTN 092), adults without HIV were enrolled at six university-affiliated clinical research sites in the USA. Part A evaluated escalating single intravenous doses or subcutaneous infusion of PGT121.414.LS, in four groups: 3 mg/kg intravenous (treatment group 1; n=3), 10 mg/kg intravenous (treatment group 2; n=4), 30 mg/kg intravenous (treatment group 3; n=3), and 5 mg/kg subcutaneous (treatment group 4; n=3). Part B evaluated repeated sequential intravenous administrations of 20 mg/kg PGT121.414.LS plus 20 mg/kg VRC07-523LS (treatment group 5; n=10) and sequential subcutaneous administrations of 5 mg/kg PGT121.414.LS plus 5 mg/kg VRC07-523LS (treatment group 6; n=10) on days 0, 112, and 224. Participants in treatment groups 1 and 2 were enrolled sequentially, with participants enrolled and randomly assigned to treatment groups 3 and 4 after a review of safety data. Participants in treatment groups 5 and 6 were randomly assigned in blocks after a review of safety data from treatment groups 1-4. The primary endpoints were safety and tolerability of mAbs, serum concentrations and pharmacokinetics of mAbs, and serum neutralising activity, assessed in participants who received all scheduled product administrations. Serum concentrations of each mAb were measured via a multiplex assay, and neutralisation activity against multiple HIV viruses was measured via the TZM-bl assay. Serum concentrations were estimated via an open, two-compartment model with first-order elimination from the central compartment. This study was registered with ClinicalTrials.gov (NCT04212091) and has been completed.</p><p><strong>Findings: </strong>Between Nov 10, 2020, and Oct 5, 2021, we enrolled 33 participants without HIV: median age was 31 years (range 22-48); 19 were assigned female sex at birth and 11 were assigned male sex at birth. Three participants and four participants were sequentially assigned to treatment groups 1 and 2, respectively, and, after safety review, six participants were randomly assigned to treatment groups 3 (n=3) and 4 (n=3); after safety review, 20 participants were randomly assigned to treatment groups 5 (n=10) and 6 (n=10). Intravenous and subcutaneous infusions were safe and well tolerated, without serious adverse events or dose-limiting toxicities. Dose escalation of PGT121.414.LS from 3 mg/kg to 30 mg/kg (intravenous) resulted in a dose-proportional increase in serum concentration of PGT121.414.LS, whether administered alone or in combination with VRC07-523LS. The estimated elimination half-life of PGT121.414.LS was 71 days (95% CI 66-75), three times that of its parental form, PGT121. The estimated subcutaneous (vs intraveno
{"title":"Safety, tolerability, pharmacokinetics, and neutralisation activities of the anti-HIV-1 monoclonal antibody PGT121.414.LS administered alone and in combination with VRC07-523LS in adults without HIV in the USA (HVTN 136/HPTN 092): a first-in-human, open-label, randomised controlled phase 1 trial.","authors":"Srilatha Edupuganti, Christopher B Hurt, Kathryn E Stephenson, Yunda Huang, Carmen A Paez, Chenchen Yu, Catherine Yen, Brett Hanscom, Zonglin He, Maurine D Miner, Theresa Gamble, Jack Heptinstall, Kelly E Seaton, Elizabeth Domin, Bob C Lin, Krisha McKee, Nicole Doria-Rose, Stephanie Regenold, Hans Spiegel, Maija Anderson, Nadia McClosky, Lily Zhang, Estelle Piwowar-Manning, Margaret E Ackerman, Michael Pensiero, Bonnie J Dye, Raphael J Landovitz, Kenneth Mayer, Marc Siegel, Magdalena Sobieszczyk, Stephen R Walsh, Lucio Gama, Dan H Barouch, David C Montefiori, Georgia D Tomaras","doi":"10.1016/S2352-3018(24)00247-9","DOIUrl":"10.1016/S2352-3018(24)00247-9","url":null,"abstract":"<p><strong>Background: </strong>Multiple broadly neutralising monoclonal antibodies (mAbs) are in development for HIV-1 prevention. The aim of this trial was to test the PGT121.414.LS and VRC07-523LS mAbs for safety and pharmacokinetics in adults.</p><p><strong>Methods: </strong>In this first-in-human phase 1 trial (HVTN 136/HPTN 092), adults without HIV were enrolled at six university-affiliated clinical research sites in the USA. Part A evaluated escalating single intravenous doses or subcutaneous infusion of PGT121.414.LS, in four groups: 3 mg/kg intravenous (treatment group 1; n=3), 10 mg/kg intravenous (treatment group 2; n=4), 30 mg/kg intravenous (treatment group 3; n=3), and 5 mg/kg subcutaneous (treatment group 4; n=3). Part B evaluated repeated sequential intravenous administrations of 20 mg/kg PGT121.414.LS plus 20 mg/kg VRC07-523LS (treatment group 5; n=10) and sequential subcutaneous administrations of 5 mg/kg PGT121.414.LS plus 5 mg/kg VRC07-523LS (treatment group 6; n=10) on days 0, 112, and 224. Participants in treatment groups 1 and 2 were enrolled sequentially, with participants enrolled and randomly assigned to treatment groups 3 and 4 after a review of safety data. Participants in treatment groups 5 and 6 were randomly assigned in blocks after a review of safety data from treatment groups 1-4. The primary endpoints were safety and tolerability of mAbs, serum concentrations and pharmacokinetics of mAbs, and serum neutralising activity, assessed in participants who received all scheduled product administrations. Serum concentrations of each mAb were measured via a multiplex assay, and neutralisation activity against multiple HIV viruses was measured via the TZM-bl assay. Serum concentrations were estimated via an open, two-compartment model with first-order elimination from the central compartment. This study was registered with ClinicalTrials.gov (NCT04212091) and has been completed.</p><p><strong>Findings: </strong>Between Nov 10, 2020, and Oct 5, 2021, we enrolled 33 participants without HIV: median age was 31 years (range 22-48); 19 were assigned female sex at birth and 11 were assigned male sex at birth. Three participants and four participants were sequentially assigned to treatment groups 1 and 2, respectively, and, after safety review, six participants were randomly assigned to treatment groups 3 (n=3) and 4 (n=3); after safety review, 20 participants were randomly assigned to treatment groups 5 (n=10) and 6 (n=10). Intravenous and subcutaneous infusions were safe and well tolerated, without serious adverse events or dose-limiting toxicities. Dose escalation of PGT121.414.LS from 3 mg/kg to 30 mg/kg (intravenous) resulted in a dose-proportional increase in serum concentration of PGT121.414.LS, whether administered alone or in combination with VRC07-523LS. The estimated elimination half-life of PGT121.414.LS was 71 days (95% CI 66-75), three times that of its parental form, PGT121. The estimated subcutaneous (vs intraveno","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":"e13-e25"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142819770","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-10-25DOI: 10.1016/S2352-3018(24)00234-0
Jane K Maganga, Khanh Pham, John M Changalucha, Jennifer A Downs
The sex of people living with HIV-1 infection, schistosome infection, or both, is a fundamental determinant of their clinical outcomes and of how these two infections interact in the host. Data from longitudinal and cross-sectional human studies and animal models indicate that males with HIV-1 and schistosome co-infection excrete fewer schistosome eggs and might have higher HIV-1 RNA viral loads and greater liver damage. Females with schistosome infection appear to have higher risk of HIV-1 acquisition than females without, particularly in Schistosoma haematobium infection, and a greater risk of death in HIV-1 and schistosome co-infection. Greater transmission of HIV-1 to partners has been shown in both sexes in those with schistosome infection. Biological sex is a consequential factor affecting pathophysiological and clinical responses in HIV-1 and schistosome co-infection. Designing future analyses to incorporate sex is vital to optimise research and care for people living with HIV-1, schistosomes, and HIV-1 and schistosome co-infection.
{"title":"Sex as a biological variable in HIV-1 and schistosome co-infection.","authors":"Jane K Maganga, Khanh Pham, John M Changalucha, Jennifer A Downs","doi":"10.1016/S2352-3018(24)00234-0","DOIUrl":"10.1016/S2352-3018(24)00234-0","url":null,"abstract":"<p><p>The sex of people living with HIV-1 infection, schistosome infection, or both, is a fundamental determinant of their clinical outcomes and of how these two infections interact in the host. Data from longitudinal and cross-sectional human studies and animal models indicate that males with HIV-1 and schistosome co-infection excrete fewer schistosome eggs and might have higher HIV-1 RNA viral loads and greater liver damage. Females with schistosome infection appear to have higher risk of HIV-1 acquisition than females without, particularly in Schistosoma haematobium infection, and a greater risk of death in HIV-1 and schistosome co-infection. Greater transmission of HIV-1 to partners has been shown in both sexes in those with schistosome infection. Biological sex is a consequential factor affecting pathophysiological and clinical responses in HIV-1 and schistosome co-infection. Designing future analyses to incorporate sex is vital to optimise research and care for people living with HIV-1, schistosomes, and HIV-1 and schistosome co-infection.</p>","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":"e60-e70"},"PeriodicalIF":12.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11710965/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142570253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-13DOI: 10.1016/S2352-3018(24)00309-6
Tanuja N Gengiah, Quarraisha Abdool Karim
{"title":"Preventing HIV in young women in Africa.","authors":"Tanuja N Gengiah, Quarraisha Abdool Karim","doi":"10.1016/S2352-3018(24)00309-6","DOIUrl":"https://doi.org/10.1016/S2352-3018(24)00309-6","url":null,"abstract":"","PeriodicalId":48725,"journal":{"name":"Lancet Hiv","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2024-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142839951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}