1
|
Kamphuis AEM, Bamford A, Tagarro A, Cressey TR, Bekker A, Amuge P, Mujuru HA, Ndongo FA, Diack A, Compagnucci A, Lallemant M, Colbers A, Turkova A. Optimising Paediatric HIV Treatment: Recent Developments and Future Directions. Paediatr Drugs 2024; 26:631-648. [PMID: 39436531 PMCID: PMC11519159 DOI: 10.1007/s40272-024-00656-4] [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] [Accepted: 09/10/2024] [Indexed: 10/23/2024]
Abstract
Treatment options for children living with HIV have historically been less effective, less practical and more difficult to implement compared with those for adults, as the research and development of new drugs for children has lagged behind. Significant progress has been achieved in response to the paediatric HIV epidemic over the last decade. Several optimised paediatric antiretroviral formulations are currently available or in development, including fixed-dose combination tablets containing a complete World Health Organization-recommended regimen. Despite these advancements, virological suppression rates in children are generally lower than in adults. Even when oral fixed-dose combinations with the optimal target profiles are developed, for some children virological suppression is not achievable for reasons such as adherence challenges, intolerance, toxicity and genotypic resistance. New safe, effective, well-tolerated antiretroviral agents from existing and novel classes, as well as innovative administration strategies are essential. To achieve the UNAIDS target of virological suppression in 95% of children receiving antiretroviral therapy, concerted efforts are required. This includes identifying priority drugs in line with latest developments, focusing drug development studies on these priorities, ensuring a timely technical knowledge transfer between originator and generic companies, accelerating regulatory approvals and facilitating procurement and implementation in countries. Success in these efforts depends on collaboration among all stakeholders, including communities, researchers, pharmaceutical companies, guideline and policymakers, governments, funders, regulators and healthcare providers. This review outlines which paediatric antiretroviral therapies are currently available, those which are under development and the future directions of paediatric HIV treatment.
Collapse
Affiliation(s)
- Anne E M Kamphuis
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Alasdair Bamford
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Medical Research Council Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, UK
| | - Alfredo Tagarro
- Fundación de Investigación Biomédica Hospital 12 de Octubre, Instituto de Investigación 12 de Octubre (imas12), Madrid, Spain
- Department of Pediatrics, Infanta Sofía University Hospital, Fundación para la Investigación Biomédica e Innovación, Hospital Universitario Infanta Sofía y Hospital del Henares (FIIB HUIS HHEN), Madrid, Spain
- Universidad Europea de Madrid, Madrid, Spain
| | - Tim R Cressey
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Adrie Bekker
- Family Centre for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch, South Africa
| | - Pauline Amuge
- Baylor College of Medicine Children's Foundation-Uganda, Kampala, Uganda
- Joint Clinical Research Centre, Kampala, Uganda
| | - Hilda Angela Mujuru
- Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | | | - Aminata Diack
- Pediatric HIV Care Unit, Centre Hospitalier National d'enfants Albert Royer, Dakar Réseau EVA, Dakar, Senegal
| | - Alexandra Compagnucci
- French National Institute of Health and Medical Research (INSERMSC10-US19-Clinical trials and Infectious Diseases), Villejuif, Paris, France
| | - Marc Lallemant
- AMS-PHPT Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Angela Colbers
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna Turkova
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Medical Research Council Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, UK
| |
Collapse
|
2
|
Cockbain B, Fidler S, Lyall H. Preventing perinatal HIV acquisition; current gaps and future perspectives. Curr Opin HIV AIDS 2024; 19:01222929-990000000-00110. [PMID: 39196368 PMCID: PMC11451969 DOI: 10.1097/coh.0000000000000881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
PURPOSE OF REVIEW Although current treatment could eradicate vertical transmission, in 2022, 130 000 infants acquired HIV globally. HIV suppression with antiretroviral therapy (ART) transforms survival for people living with HIV (PLWH), and prevents transmission, including vertical. International guidelines recommend lifelong ART for PLWH, consequently perinatal HIV acquisition reflects implementation gaps in the HIV care cascade. We summarize these gaps, exploring potential novel approaches and therapeutic innovations towards eliminating vertical HIV transmission. RECENT FINDINGS Multifactorial challenges continue to underpin gaps in the HIV care cascade, including accessibility, availability and sustainability of HIV testing, prevention and treatment, alongside stigma, gender-based violence and poverty. Long-acting ART may be important in preventing perinatal HIV acquisition, with early data demonstrating tolerability and efficacy of injectable ART throughout pregnancy, both as HIV treatment and prevention. Carefully selected long-acting broadly neutralizing antibodies (bNAbs) matching circulating, exposing viral envelope sequences have demonstrated safety, clinical trials are ongoing to demonstrate efficacy. SUMMARY Emerging clinical studies should prioritize pregnant/lactating people and infants to ensure such therapies are well tolerated and efficacious. Alongside therapeutic innovation, programmatic strategies must address social and economic challenges, ensuring sustainable HIV treatment/prevention programmes and facilitating global elimination of blood-borne viruses.
Collapse
Affiliation(s)
- Beatrice Cockbain
- Department of Infectious Disease, Imperial College London, Imperial College NIHR BRC
- Chelsea and Westminster Hospital NHS Foundation Trust
| | - Sarah Fidler
- Department of Infectious Disease, Imperial College London, Imperial College NIHR BRC
- Department of Infectious Disease and NIHR Imperial BRC, Imperial College London, UK
| | - Hermione Lyall
- Department of Infectious Disease and NIHR Imperial BRC, Imperial College London, UK
| |
Collapse
|
3
|
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
|
4
|
Mahomed S. Broadly neutralizing antibodies for HIV prevention: a comprehensive review and future perspectives. Clin Microbiol Rev 2024; 37:e0015222. [PMID: 38687039 PMCID: PMC11324036 DOI: 10.1128/cmr.00152-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
SUMMARYThe human immunodeficiency virus (HIV) epidemic remains a formidable global health concern, with 39 million people living with the virus and 1.3 million new infections reported in 2022. Despite anti-retroviral therapy's effectiveness in pre-exposure prophylaxis, its global adoption is limited. Broadly neutralizing antibodies (bNAbs) offer an alternative strategy for HIV prevention through passive immunization. Historically, passive immunization has been efficacious in the treatment of various diseases ranging from oncology to infectious diseases. Early clinical trials suggest bNAbs are safe, tolerable, and capable of reducing HIV RNA levels. Although challenges such as bNAb resistance have been noted in phase I trials, ongoing research aims to assess the additive or synergistic benefits of combining multiple bNAbs. Researchers are exploring bispecific and trispecific antibodies, and fragment crystallizable region modifications to augment antibody efficacy and half-life. Moreover, the potential of other antibody isotypes like IgG3 and IgA is under investigation. While promising, the application of bNAbs faces economic and logistical barriers. High manufacturing costs, particularly in resource-limited settings, and logistical challenges like cold-chain requirements pose obstacles. Preliminary studies suggest cost-effectiveness, although this is contingent on various factors like efficacy and distribution. Technological advancements and strategic partnerships may mitigate some challenges, but issues like molecular aggregation remain. The World Health Organization has provided preferred product characteristics for bNAbs, focusing on optimizing their efficacy, safety, and accessibility. The integration of bNAbs in HIV prophylaxis necessitates a multi-faceted approach, considering economic, logistical, and scientific variables. This review comprehensively covers the historical context, current advancements, and future avenues of bNAbs in HIV prevention.
Collapse
Affiliation(s)
- Sharana Mahomed
- Centre for the AIDS
Programme of Research in South Africa (CAPRISA), Doris Duke Medical
Research Institute, Nelson R Mandela School of Medicine, University of
KwaZulu-Natal, Durban,
South Africa
| |
Collapse
|
5
|
Yin DE, Palin AC, Lombo TB, Mahon RN, Poon B, Wu DY, Atala A, Brooks KM, Chen S, Coyne CB, D’Souza MP, Fackler OT, Furler O’Brien RL, Garcia-de-Alba C, Jean-Philippe P, Karn J, Majji S, Muotri AR, Ozulumba T, Sakatis MZ, Schlesinger LS, Singh A, Spiegel HM, Struble E, Sung K, Tagle DA, Thacker VV, Tidball AM, Varthakavi V, Vunjak-Novakovic G, Wagar LE, Yeung CK, Ndhlovu LC, Ott M. 3D human tissue models and microphysiological systems for HIV and related comorbidities. Trends Biotechnol 2024; 42:526-543. [PMID: 38071144 PMCID: PMC11065605 DOI: 10.1016/j.tibtech.2023.10.008] [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: 07/03/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 03/03/2024]
Abstract
Three-dimensional (3D) human tissue models/microphysiological systems (e.g., organs-on-chips, organoids, and tissue explants) model HIV and related comorbidities and have potential to address critical questions, including characterization of viral reservoirs, insufficient innate and adaptive immune responses, biomarker discovery and evaluation, medical complexity with comorbidities (e.g., tuberculosis and SARS-CoV-2), and protection and transmission during pregnancy and birth. Composed of multiple primary or stem cell-derived cell types organized in a dedicated 3D space, these systems hold unique promise for better reproducing human physiology, advancing therapeutic development, and bridging the human-animal model translational gap. Here, we discuss the promises and achievements with 3D human tissue models in HIV and comorbidity research, along with remaining barriers with respect to cell biology, virology, immunology, and regulatory issues.
Collapse
|
6
|
Sakoi-Mosetlhi M, Ajibola G, Haghighat R, Batlang O, Maswabi K, Pretorius-Holme M, Powis KM, Lockman S, Makhema J, Litcherfeld M, Kuritzkes DR, Shapiro R. Caregivers of children with HIV in Botswana prefer monthly IV Broadly Neutralizing Antibodies (bNAbs) to daily oral ART. PLoS One 2024; 19:e0299942. [PMID: 38536810 PMCID: PMC10971757 DOI: 10.1371/journal.pone.0299942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/20/2024] [Indexed: 04/01/2024] Open
Abstract
INTRODUCTION Monthly intravenous infusion of broadly neutralizing monoclonal antibodies may be an attractive alternative to daily oral antiretroviral treatment for children living with HIV. However, acceptability among caregivers remains unknown. METHODS We evaluated monthly infusion of dual bNAbs (VRCO1LS and 10-1074) as a treatment alternative to ART among children participating in the Tatelo Study in Botswana. Eligible children aged 2-5 years received 8-32 weeks of bNAbs overlapping with ART, and up to 24 weeks of bNAbs alone as monthly intravenous infusion. Using closed-ended questionnaires, we evaluated caregiver acceptability of each treatment strategy prior to the first bNAb administration visit (pre-intervention) and after the completion of the final bNAb administration visit (post-intervention). RESULTS Twenty-five children completed the intervention phase of the study, and acceptability data were available from 24 caregivers at both time points. Responses were provided by the child's mother at both visits (60%), an extended family member at both visits (28%), or a combination of mother and an extended family member (12%). Caregiver acceptance of monthly bNAb infusions was extremely high both pre-and post-intervention, with 21/24 (87.5%) preferring bNAbs to ART pre-intervention, and 21/25 (84%) preferring bNAbs post-intervention. While no caregiver preferred ART pre-intervention, 2/25 preferred it post-intervention. Pre-intervention, 3 (13%) caregivers had no preference between monthly bNAbs or daily ART, and 2 (8%) had no preference post-intervention. Pre-intervention, the most common reasons for preferring bNAbs over ART were the perception that bNAbs were better at suppressing the virus than ART (n = 10) and the fact that infusions were dosed once monthly compared to daily ART (n = 9). Post-intervention, no dominant reason for preferring bNAbs over ART emerged from caregivers. CONCLUSIONS Monthly intravenous bNAb infusions were highly acceptable to caregivers of children with HIV in Botswana and preferred over standard ART by the majority of caregivers. CLINICAL TRIAL NUMBER NCT03707977.
Collapse
Affiliation(s)
| | | | - Roxanna Haghighat
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Oganne Batlang
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Kenneth Maswabi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Molly Pretorius-Holme
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Kathleen M. Powis
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Departments of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Shahin Lockman
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Joseph Makhema
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Mathias Litcherfeld
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Daniel R. Kuritzkes
- Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Roger Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
7
|
Van de Perre P, Scarlatti G, Moore PL, Molès JP, Nagot N, Tylleskär T, Gray G, Goga A. Preventing breast milk HIV transmission using broadly neutralizing monoclonal antibodies: One size does not fit all. Immun Inflamm Dis 2024; 12:e1216. [PMID: 38533917 DOI: 10.1002/iid3.1216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Key messages
Passive immunoprophylaxis with broadly neutralizing monoclonal antibodies (bNAbs) could be a game changer in the prevention of human immunodeficiency virus (HIV) acquisition.
The prevailing view is that available resources should be focused on identifying a fixed combination of at least three bNAbs for universal use in therapeutic and preventive protocols, regardless of target populations or routes of transmission.
HIV transmission through breastfeeding is unique: it involves free viral particles and cell‐associated virus from breast milk and, in the case of acute/recent maternal infection, a viral population with restricted Env diversity.
HIV transmission through breastfeeding in high incidence/prevalence areas could potentially be eliminated by subcutaneous administration to all newborns of one or two long‐acting bNAbs with extended breadth, high potency, and effector properties (ADCC, phagocytosis) against circulating HIV strains.
Collapse
Affiliation(s)
- Philippe Van de Perre
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Etablissement Français du Sang, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Penny L Moore
- MRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Jean-Pierre Molès
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Etablissement Français du Sang, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Nicolas Nagot
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Etablissement Français du Sang, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Thorkild Tylleskär
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Glenda Gray
- South African Medical Research Council, Cape Town, South Africa
| | - Ameena Goga
- South African Medical Research Council, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
8
|
Bamford A, Foster C, Lyall H. Infant feeding: emerging concepts to prevent HIV transmission. Curr Opin Infect Dis 2024; 37:8-16. [PMID: 37889586 PMCID: PMC10734781 DOI: 10.1097/qco.0000000000000986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
PURPOSE OF REVIEW HIV screening in pregnancy, universal suppressive antiretroviral therapy (ART) and breastfeeding avoidance can almost completely prevent vertical transmission of HIV. Breastfeeding is associated with an additional risk of transmission, although this risk is extremely low with suppressive maternal ART. This minimal risk must be balanced with the benefits of breastfeeding for women living with HIV (WLHIV) and their infants. Guidance in high-income countries has evolved, moving towards supported breast feeding for women on suppressive ART. RECENT FINDINGS Breastmilk transmission accounts for an increasing proportion of new infant infections globally. The majority of transmission data comes from studies including women not on suppressive ART. Breastmilk transmissions in the context of undetectable viral load have rarely occurred, although risk factors remain unclear. Outcome data on supported breastfeeding are accumulating, providing evidence for guidelines and informing infant feeding decisions. Long-acting ART for maternal preexposure prophylaxis or treatment, and infant postnatal prophylaxis are promising future options. SUMMARY Breastfeeding on suppressive ART has a very low risk of vertical transmission and can have multiple benefits for WLHIV and their infants. However, caution is advised with relaxation of breastfeeding guidance so as not to jeopardise the global goal of elimination of vertical transmission by 2030.
Collapse
Affiliation(s)
- Alasdair Bamford
- Great Ormond Street Hospital for Children NHS Foundation Trust
- UCL Great Ormond Street Institute of Child Health
- MRC Clinical Trials Unit at UCL
| | - Caroline Foster
- Imperial College Healthcare NHS Trust, St Mary's Hospital, London, UK
| | - Hermione Lyall
- Imperial College Healthcare NHS Trust, St Mary's Hospital, London, UK
| |
Collapse
|
9
|
Sliefert M, Maloba M, Wexler C, Were F, Mbithi Y, Mugendi G, Maliski E, Nicolay Z, Thomas G, Kale S, Maosa N, Finocchario-Kessler S. Challenges with pediatric antiretroviral therapy administration: Qualitative perspectives from caregivers and HIV providers in Kenya. PLoS One 2024; 19:e0296713. [PMID: 38194419 PMCID: PMC10775971 DOI: 10.1371/journal.pone.0296713] [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: 05/16/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Current formulations of pediatric antiretroviral therapy (ART) for children with HIV present significant barriers to adherence, leading to drug resistance, ART ineffectiveness, and preventable child morbidity and mortality. Understanding these challenges and how they contribute to suboptimal adherence is an important step in improving outcomes. This qualitative study describes how regimen-related challenges create barriers to adherence and impact families. METHODS We conducted key informant interviews (KIIs) with 30 healthcare providers and 9 focus group discussions (FGDs) with a total of 72 caregivers, across three public hospitals in Siaya and Mombasa Kenya. The KIIs and FGDs were audio recorded, translated, and transcribed verbatim. The transcripts were hand coded based on emergent and a-priori themes. RESULTS Caregivers discussed major regimen-related challenges to adherence included poor palatability of current formulations, complex preparation, and administration (including measuring, crushing, dissolving, mixing), complex drug storage, and frequent refill appointments and how these regimen-related challenges contributed to individual and intrapersonal barriers to adherence. Caregivers discussed how poor taste led to child anxiety, refusal of medications, and the need for caregivers to use bribes or threats during administration. Complex preparation led to concerns and challenges about maintaining privacy and confidentiality, especially during times of travel. Providers corroborated this patient experience and described how these challenges with administration led to poor infant outcomes, including high viral load and preventable morbidity. Providers discussed how the frequency of refills could range from every 2 weeks to every 3 months, depending on the patient. Caregivers discussed how these refill frequencies interrupted work and school schedules, risked unwanted disclosure to peers, required use of financial resources for travel, and ultimately were a challenge to adherence. CONCLUSION These findings highlight the need for improved formulations for pediatric ART to ease the daily burden on caregivers and children to increase adherence, improve child health, and overall quality of life of families.
Collapse
Affiliation(s)
- Michala Sliefert
- University of Kansas Medical Center, Kansas City, KS, United States of America
| | - May Maloba
- Global Health Innovations, Nairobi, Kenya
| | - Catherine Wexler
- University of Kansas Medical Center, Kansas City, KS, United States of America
| | | | | | | | - Edward Maliski
- Oak Therapeutics, Lawrence, KS, United States of America
| | | | - Gregory Thomas
- University of Kansas, Lawrence, KS, United States of America
| | | | | | | |
Collapse
|
10
|
Joseph J, Sandel G, Kulkarni R, Alatrash R, Herrera BB, Jain P. Antibody and Cell-Based Therapies against Virus-Induced Cancers in the Context of HIV/AIDS. Pathogens 2023; 13:14. [PMID: 38251321 PMCID: PMC10821063 DOI: 10.3390/pathogens13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Infectious agents, notably viruses, can cause or increase the risk of cancer occurrences. These agents often disrupt normal cellular functions, promote uncontrolled proliferation and growth, and trigger chronic inflammation, leading to cancer. Approximately 20% of all cancer cases in humans are associated with an infectious pathogen. The International Agency for Research on Cancer (IARC) recognizes seven viruses as direct oncogenic agents, including Epstein-Barr Virus (EBV), Kaposi's Sarcoma-associated herpesvirus (KSHV), human T-cell leukemia virus type-1 (HTLV-1), human papilloma virus (HPV), hepatitis C virus (HCV), hepatitis B virus (HBV), and human immunodeficiency virus type 1 (HIV-1). Most viruses linked to increased cancer risk are typically transmitted through contact with contaminated body fluids and high-risk behaviors. The risk of infection can be reduced through vaccinations and routine testing, as well as recognizing and addressing risky behaviors and staying informed about public health concerns. Numerous strategies are currently in pre-clinical phases or undergoing clinical trials for targeting cancers driven by viral infections. Herein, we provide an overview of risk factors associated with increased cancer incidence in people living with HIV (PLWH) as well as other chronic viral infections, and contributing factors such as aging, toxicity from ART, coinfections, and comorbidities. Furthermore, we highlight both antibody- and cell-based strategies directed against virus-induced cancers while also emphasizing approaches aimed at discovering cures or achieving complete remission for affected individuals.
Collapse
Affiliation(s)
- Julie Joseph
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Grace Sandel
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Ratuja Kulkarni
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Reem Alatrash
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Bobby Brooke Herrera
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Pooja Jain
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| |
Collapse
|
11
|
Alba C, Malhotra S, Horsfall S, Barnhart ME, Bekker A, Chapman K, Cunningham CK, Fast PE, Fouda GG, Freedberg KA, Goga A, Ghazaryan LR, Leroy V, Mann C, McCluskey MM, McFarland EJ, Muturi-Kioi V, Permar SR, Shapiro R, Sok D, Stranix-Chibanda L, Weinstein MC, Ciaranello AL, Dugdale CM. Cost-effectiveness of broadly neutralizing antibodies for infant HIV prophylaxis in settings with high HIV burdens: a simulation modeling study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.06.23298184. [PMID: 37986879 PMCID: PMC10659508 DOI: 10.1101/2023.11.06.23298184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Introduction Approximately 130 000 infants acquire HIV annually despite global maternal antiretroviral therapy scale-up. We evaluated the potential clinical impact and cost-effectiveness of offering long-acting, anti-HIV broadly neutralizing antibody (bNAb) prophylaxis to infants in three distinct settings. Methods We simulated infants in Côte d'Ivoire, South Africa, and Zimbabwe using the Cost-Effectiveness of Preventing AIDS Complications-Pediatric (CEPAC-P) model. We modeled strategies offering a three-bNAb combination in addition to WHO-recommended standard-of-care oral prophylaxis to infants: a) with known, WHO-defined high-risk HIV exposure at birth (HR-HIVE); b) with known HIV exposure at birth (HIVE); or c) with or without known HIV exposure (ALL). Modeled infants received 1-dose, 2-doses, or Extended (every 3 months through 18 months) bNAb dosing. Base case model inputs included 70% bNAb efficacy (sensitivity analysis range: 10-100%), 3-month efficacy duration/dosing interval (1-6 months), and $20/dose cost ($5-$100/dose). Outcomes included pediatric HIV infections, life expectancy, lifetime HIV-related costs, and incremental cost-effectiveness ratios (ICERs, in US$/year-of-life-saved [YLS], assuming a ≤50% GDP per capita cost-effectiveness threshold). Results The base case model projects that bNAb strategies targeting HIVE and ALL infants would prevent 7-26% and 10-42% additional pediatric HIV infections, respectively, compared to standard-of-care alone, ranging by dosing approach. HIVE-Extended would be cost-effective (cost-saving compared to standard-of-care) in Côte d'Ivoire and Zimbabwe; ALL-Extended would be cost-effective in South Africa (ICER: $882/YLS). BNAb strategies targeting HR-HIVE infants would result in greater lifetime costs and smaller life expectancy gains than HIVE-Extended. Throughout most bNAb efficacies and costs evaluated in sensitivity analyses, targeting HIVE infants would be cost-effective in Côte d'Ivoire and Zimbabwe, and targeting ALL infants would be cost-effective in South Africa. Discussion Adding long-acting bNAbs to current standard-of-care prophylaxis would be cost-effective, assuming plausible efficacies and costs. The cost-effective target population would vary by setting, largely driven by maternal antenatal HIV prevalence and postpartum incidence.
Collapse
Affiliation(s)
- Christopher Alba
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, United States
| | | | - Stephanie Horsfall
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, United States
| | - Matthew E. Barnhart
- Office of HIV/AIDS, Bureau for Global Health, Agency for International Development (USAID), District of Columbia, United States
| | - Adrie Bekker
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | - Coleen K. Cunningham
- Department of Pediatrics, University of California Irvine, Irvine, United States
- Department of Pediatrics, Children’s Hospital of Orange County, Orange, United States
| | - Patricia E. Fast
- IAVI, New York, United States
- Pediatric Infectious Diseases, Stanford University School of Medicine, Palo Alto, United States
| | - Genevieve G. Fouda
- Department of Pediatrics, New York-Presbyterian/Weill Cornell Medical Center, New York, United States
| | - Kenneth A. Freedberg
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, United States
- Harvard Medical School, Boston, United States
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, United States
- Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Ameena Goga
- South African Medical Research Council, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| | - Lusine R. Ghazaryan
- Office of HIV/AIDS, Bureau for Global Health, Agency for International Development (USAID), District of Columbia, United States
| | - Valériane Leroy
- Centre d’Epidémiologie et de Recherche en santé des POPulations (CERPOP), Inserm and Université Toulouse III, Toulouse, France
| | - Carlyn Mann
- Office of HIV/AIDS, Bureau for Global Health, Agency for International Development (USAID), District of Columbia, United States
| | - Margaret M. McCluskey
- Office of HIV/AIDS, Bureau for Global Health, Agency for International Development (USAID), District of Columbia, United States
| | - Elizabeth J. McFarland
- Department of Pediatrics, Children’s Hospital Colorado and University of Colorado Anschutz Medical Campus, Aurora, United States
| | | | - Sallie R. Permar
- Department of Pediatrics, New York-Presbyterian/Weill Cornell Medical Center, New York, United States
| | - Roger Shapiro
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, United States
| | - Devin Sok
- IAVI, New York, United States
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, United States
| | - Lynda Stranix-Chibanda
- Child and Adolescent Health Unit, University of Zimbabwe Faculty of Medicine and Health Sciences, Harare, Zimbabwe
| | - Milton C. Weinstein
- Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Andrea L. Ciaranello
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, United States
- Harvard Medical School, Boston, United States
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, United States
| | - Caitlin M. Dugdale
- Medical Practice Evaluation Center, Massachusetts General Hospital, Boston, United States
- Harvard Medical School, Boston, United States
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, United States
| |
Collapse
|
12
|
Shapiro RL, Ajibola G, Maswabi K, Hughes M, Nelson BS, Niesar A, Holme MP, Powis KM, Sakoi M, Batlang O, Moyo S, Mohammed T, Maphorisa C, Bennett K, Hu Z, Giguel F, Reeves JD, Reeves MA, Gao C, Yu X, Ackerman ME, McDermott A, Cooper M, Caskey M, Gama L, Jean-Philippe P, Yin DE, Capparelli EV, Lockman S, Makhema J, Kuritzkes DR, Lichterfeld M. Broadly neutralizing antibody treatment maintained HIV suppression in children with favorable reservoir characteristics in Botswana. Sci Transl Med 2023; 15:eadh0004. [PMID: 37406137 PMCID: PMC10683791 DOI: 10.1126/scitranslmed.adh0004] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/26/2023] [Indexed: 07/07/2023]
Abstract
Broadly neutralizing antibodies (bNAbs) may provide an alternative to standard antiretroviral treatment (ART) for controlling HIV-1 replication and may have immunotherapeutic effects against HIV-1 reservoirs. We conducted a prospective clinical trial with two HIV-1 bNAbs (VRC01LS and 10-1074) in children (n = 25) who had previously initiated small-molecule ART treatment before 7 days of age and who continued treatment for at least 96 weeks. Both bNAbs were dosed intravenously every 4 weeks, overlapping with ART for at least 8 weeks and then continued for up to 24 weeks or until detectable viremia of HIV-1 RNA rose above 400 copies per milliliter in the absence of ART. Eleven (44%) children maintained HIV-1 RNA below 400 copies per milliliter through 24 weeks of bNAb-only treatment; 14 (56%) had detectable viremia above 400 copies per milliliter at a median of 4 weeks. Archived HIV-1 provirus susceptible to 10-1074, lower birth HIV-1 DNA reservoir in peripheral blood mononuclear cells, sustained viral suppression throughout early life, and combined negative qualitative HIV-1 DNA polymerase chain reaction and negative HIV-1 serology at entry were associated with maintaining suppression on bNAbs alone. This proof-of-concept study suggests that bNAbs may represent a promising treatment modality for infants and children living with HIV-1. Future studies using newer bNAb combinations with greater breadth and potency are warranted.
Collapse
Affiliation(s)
- Roger L. Shapiro
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Botswana Harvard Health Partnership; Gaborone, Botswana
| | | | | | - Michael Hughes
- Department of Biostatistics, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
| | - Bryan S. Nelson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
| | - Aischa Niesar
- Ragon Institute of MGH, MIT and Harvard; Cambridge, MA 02139, USA
| | - Molly Pretorius Holme
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
| | - Kathleen M. Powis
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Botswana Harvard Health Partnership; Gaborone, Botswana
- Departments of Internal Medicine and Pediatrics, Massachusetts General Hospital; Boston, MA 02114, USA
| | - Maureen Sakoi
- Botswana Harvard Health Partnership; Gaborone, Botswana
| | | | - Sikhulile Moyo
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Botswana Harvard Health Partnership; Gaborone, Botswana
| | | | | | - Kara Bennett
- Bennett Statistical Consulting, Inc.; Ballston Lake, NY 12019, USA
| | - Zixin Hu
- Division of Infectious Diseases, Brigham and Women’s Hospital; Boston, MA 02115, USA
| | - Francoise Giguel
- Division of Infectious Diseases, Brigham and Women’s Hospital; Boston, MA 02115, USA
| | | | - Michael A. Reeves
- Labcorp-Monogram Biosciences, Inc.; South San Francisco, CA 94080, USA
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard; Cambridge, MA 02139, USA
| | - Xu Yu
- Ragon Institute of MGH, MIT and Harvard; Cambridge, MA 02139, USA
| | | | | | - Marlene Cooper
- Frontier Science and Technology Research Foundation, Inc.; Amherst, NY 14226, USA
| | | | - Lucio Gama
- Vaccine Research Center; Bethesda, MD 20892, USA
| | - Patrick Jean-Philippe
- National Institute of Allergy and Infectious Diseases, National Institutes of Health; Rockville, MD 20892, USA
| | - Dwight E. Yin
- National Institute of Allergy and Infectious Diseases, National Institutes of Health; Rockville, MD 20892, USA
| | - Edmund V. Capparelli
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92037, USA
| | - Shahin Lockman
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Botswana Harvard Health Partnership; Gaborone, Botswana
- Division of Infectious Diseases, Brigham and Women’s Hospital; Boston, MA 02115, USA
| | - Joseph Makhema
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Botswana Harvard Health Partnership; Gaborone, Botswana
| | - Daniel R. Kuritzkes
- Division of Infectious Diseases, Brigham and Women’s Hospital; Boston, MA 02115, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard; Cambridge, MA 02139, USA
- Division of Infectious Diseases, Brigham and Women’s Hospital; Boston, MA 02115, USA
| |
Collapse
|
13
|
Seaton KE, Huang Y, Karuna S, Heptinstall JR, Brackett C, Chiong K, Zhang L, Yates NL, Sampson M, Rudnicki E, Juraska M, deCamp AC, Edlefsen PT, Mullins JI, Williamson C, Rossenkhan R, Giorgi EE, Kenny A, Angier H, Randhawa A, Weiner JA, Rojas M, Sarzotti-Kelsoe M, Zhang L, Sawant S, Ackerman ME, McDermott AB, Mascola JR, Hural J, McElrath MJ, Andrew P, Hidalgo JA, Clark J, Laher F, Orrell C, Frank I, Gonzales P, Edupuganti S, Mgodi N, Corey L, Morris L, Montefiori D, Cohen MS, Gilbert PB, Tomaras GD. Pharmacokinetic serum concentrations of VRC01 correlate with prevention of HIV-1 acquisition. EBioMedicine 2023; 93:104590. [PMID: 37300931 PMCID: PMC10363420 DOI: 10.1016/j.ebiom.2023.104590] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND The phase 2b proof-of-concept Antibody Mediated Prevention (AMP) trials showed that VRC01, an anti-HIV-1 broadly neutralising antibody (bnAb), prevented acquisition of HIV-1 sensitive to VRC01. To inform future study design and dosing regimen selection of candidate bnAbs, we investigated the association of VRC01 serum concentration with HIV-1 acquisition using AMP trial data. METHODS The case-control sample included 107 VRC01 recipients who acquired HIV-1 and 82 VRC01 recipients who remained without HIV-1 during the study. We measured VRC01 serum concentrations with a qualified pharmacokinetic (PK) Binding Antibody Multiplex Assay. We employed nonlinear mixed effects PK modelling to estimate daily-grid VRC01 concentrations. Cox regression models were used to assess the association of VRC01 concentration at exposure and baseline body weight, with the hazard of HIV-1 acquisition and prevention efficacy as a function of VRC01 concentration. We also compared fixed dosing vs. body weight-based dosing via simulations. FINDINGS Estimated VRC01 concentrations in VRC01 recipients without HIV-1 were higher than those in VRC01 recipients who acquired HIV-1. Body weight was inversely associated with HIV-1 acquisition among both placebo and VRC01 recipients but did not modify the prevention efficacy of VRC01. VRC01 concentration was inversely correlated with HIV-1 acquisition, and positively correlated with prevention efficacy of VRC01. Simulation studies suggest that fixed dosing may be comparable to weight-based dosing in overall predicted prevention efficacy. INTERPRETATION These findings suggest that bnAb serum concentration may be a useful marker for dosing regimen selection, and operationally efficient fixed dosing regimens could be considered for future trials of HIV-1 bnAbs. FUNDING Was provided by the National Institutes of Health, National Institute of Allergy and Infectious Diseases (NIAID) (UM1 AI068614, to the HIV Vaccine Trials Network [HVTN]; UM1 AI068635, to the HVTN Statistical Data and Management Center [SDMC], Fred Hutchinson Cancer Center [FHCC]; 2R37 054165 to the FHCC; UM1 AI068618, to HVTN Laboratory Center, FHCC; UM1 AI068619, to the HPTN Leadership and Operations Center; UM1 AI068613, to the HIV Prevention Trials Network [HPTN] Laboratory Center; UM1 AI068617, to the HPTN SDMC; and P30 AI027757, to the Center for AIDS Research, Duke University (AI P30 AI064518) and University of Washington (P30 AI027757) Centers for AIDS Research; R37AI054165 from NIAID to the FHCC; and OPP1032144 CA-VIMC Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Kelly E Seaton
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA.
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA; Department of Global Health, University of Washington, Seattle, WA, 98195, USA.
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jack R Heptinstall
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Caroline Brackett
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Kelvin Chiong
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Lily Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Nicole L Yates
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Mark Sampson
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Erika Rudnicki
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Allan C deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James I Mullins
- Department of Global Health, University of Washington, Seattle, WA, 98195, USA; Departments of Microbiology and Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Carolyn Williamson
- Division of Medical Virology, Institute of Infectious Disease & Molecular Medicine, University of Cape Town and National Health Laboratory Service, South Africa
| | - Raabya Rossenkhan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Elena E Giorgi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Avi Kenny
- Department of Biostatistics, University of Washington, Seattle, WA, 98195, USA
| | - Heather Angier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - April Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Michelle Rojas
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Marcella Sarzotti-Kelsoe
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Lu Zhang
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Sheetal Sawant
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | | | | | | | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - M Julianna McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | | | | | - Jesse Clark
- Department of Medicine, Division of Infectious Disease and Department of Family Medicine in the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Fatima Laher
- Perinatal HIV Research Unit (PHRU), Wits Health Consortium, Soweto, Johannesburg, South Africa
| | - Catherine Orrell
- Desmond Tutu Health Foundation, University of Cape Town (Institute of Infectious Disease and Molecular Medicine, and Department of Medicine), Observatory, 7925, Cape Town, South Africa
| | - Ian Frank
- Penn Center for AIDS Research, Infectious Disease Division, University of Pennsylvania, 3400 Civic Center Boulevard Building 421, Philadelphia, PA, 19104, USA
| | - Pedro Gonzales
- Asociacion Civil Impacta Salud y Educación, San Miguel Clinical Research Center, Lima, Peru
| | - Srilatha Edupuganti
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Nyaradzo Mgodi
- University of Zimbabwe-University of California San Francisco (UZ-UCSF) Collaborative Research Programme, Harare, Zimbabwe, South Africa
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA; Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA, 98195, USA; Division of Medical Virology, University of Cape Town, Anzio Road, Observatory, 7925, Cape Town, South Africa
| | - Lynn Morris
- National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, 2192, South Africa; Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2000, South Africa; Centre for the AIDS Programme of Research in South Africa, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - David Montefiori
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Myron S Cohen
- Institute of Global Health and Infectious Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA; Departments of Microbiology and Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Georgia D Tomaras
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA.
| |
Collapse
|
14
|
Ajibola G, Masheto G, Shapiro R. Antibody interventions in HIV: broadly neutralizing mAbs in children. Curr Opin HIV AIDS 2023; 18:217-224. [PMID: 37278286 DOI: 10.1097/coh.0000000000000806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
PURPOSE OF REVIEW Treatment strategies for children with HIV are evolving, with considerations beyond plasma viremic control that raise the possibility of reducing or eliminating latent reservoirs to achieve posttreatment control. Novel strategies that maintain HIV viral suppression and allow time off small molecule antiretroviral therapy (ART) are of high priority. Trials with broadly neutralizing mAbs (bNAbs) have begun in children and may become a viable alternative treatment option. Recent bNAb treatment studies in adults indicate that bNAbs may be associated with a reduction in viral reservoirs, providing optimism that these agents may provide a pathway towards posttreatment control that rarely occurs with small molecule ART. RECENT FINDINGS Children with HIV provide an ideal opportunity to study bNAbs as an alternative treatment strategy that reduces direct ART toxicities during critical periods of growth and development, allows time off ART and takes advantage of the distinct features of the developing immune system in children that could facilitate induction of more potent autologous cellular and humoral immune responses against HIV-1. To date, paediatric bNAb studies with reported results include IMPAACT P1112, IMPAACT 2008, IMPAACT P1115 and the Tatelo study, and these results will be reviewed. SUMMARY In this review, we summarize the current and planned paediatric bNAb studies, with an emphasis on trial results available to date. We highlight the potential benefits of immune-based therapies for the maintenance of viral suppression and its potential for achieving viral remission in children living with HIV.
Collapse
Affiliation(s)
| | - Gaerolwe Masheto
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, Massachusetts, USA
| | - Roger Shapiro
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Ruel T, Penazzato M, Zech JM, Archary M, Cressey TR, Goga A, Harwell J, Landovitz RJ, Lain MG, Lallemant M, Namusoke-Magongo E, Mukui I, Permar SR, Prendergast AJ, Shapiro R, Abrams EJ. Novel Approaches to Postnatal Prophylaxis to Eliminate Vertical Transmission of HIV. GLOBAL HEALTH, SCIENCE AND PRACTICE 2023; 11:e2200401. [PMID: 37116934 PMCID: PMC10141432 DOI: 10.9745/ghsp-d-22-00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/01/2023] [Indexed: 04/03/2023]
Abstract
Despite progress in providing antiretroviral therapy to pregnant women living with HIV, a substantial number of vertical transmissions continue to occur. Novel approaches leveraging modern potent, safe, and well-tolerated antiretroviral drugs are urgently needed.
Collapse
Affiliation(s)
- Theodore Ruel
- University of California, San Francisco, San Francisco, CA, USA
| | | | - Jennifer M. Zech
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, NY, USA
| | | | - Tim R. Cressey
- AMS-IRD Research Collaboration, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Ameena Goga
- HIV and other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Pretoria, South Africa
| | | | - Raphael J. Landovitz
- UCLA Center for Clinical AIDS Research and Education, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Marc Lallemant
- AMS-PHPT Research Collaboration, Chiang Mai University, Chiang Mia, Thailand
- Penta Foundation Italy, Padova, Italy
| | | | - Irene Mukui
- Drugs for Neglected Diseases Initiative, Nairobi, Kenya
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Andrew J. Prendergast
- Queen Mary University of London, London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Roger Shapiro
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Elaine J. Abrams
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
16
|
Dugdale CM, Ufio O, Alba C, Permar SR, Stranix‐Chibanda L, Cunningham CK, Fouda GG, Myer L, Weinstein MC, Leroy V, McFarland EJ, Freedberg KA, Ciaranello AL. Cost-effectiveness of broadly neutralizing antibody prophylaxis for HIV-exposed infants in sub-Saharan African settings. J Int AIDS Soc 2023; 26:e26052. [PMID: 36604316 PMCID: PMC9816086 DOI: 10.1002/jia2.26052] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 11/28/2022] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Infant HIV prophylaxis with broadly neutralizing anti-HIV antibodies (bNAbs) could provide long-acting protection against vertical transmission. We sought to estimate the potential clinical impact and cost-effectiveness of hypothetical bNAb prophylaxis programmes for children known to be HIV exposed at birth in three sub-Saharan African settings. METHODS We conducted a cost-effectiveness analysis using the CEPAC-Pediatric model, simulating cohorts of infants from birth through death in Côte d'Ivoire, South Africa and Zimbabwe. These settings were selected to reflect a broad range of HIV care cascade characteristics, antenatal HIV prevalence and budgetary constraints. We modelled strategies targeting bNAbs to only WHO-designated "high-risk" HIV-exposed infants (HR-HIVE) or to all HIV-exposed infants (HIVE). We compared four prophylaxis approaches within each target population: standard of care oral antiretroviral prophylaxis (SOC), and SOC plus bNAbs at birth (1-dose), at birth and 3 months (2-doses), or every 3 months throughout breastfeeding (Extended). Base-case model inputs included bNAb efficacy (60%/dose), effect duration (3 months/dose) and costs ($60/dose), based on published literature. Outcomes included paediatric HIV incidence and incremental cost-effectiveness ratios (ICERs) calculated from discounted life expectancy and lifetime HIV-related costs. RESULTS The model projects that bNAbs would reduce absolute infant HIV incidence by 0.3-2.2% (9.6-34.9% relative reduction), varying by country, prophylaxis approach and target population. In all three settings, HR-HIVE-1-dose would be cost-saving compared to SOC. Using a 50% GDP per capita ICER threshold, HIVE-Extended would be cost-effective in all three settings with ICERs of $497/YLS in Côte d'Ivoire, $464/YLS in South Africa and $455/YLS in Zimbabwe. In all three settings, bNAb strategies would remain cost-effective at costs up to $200/dose if efficacy is ≥30%. If the bNAb effect duration were reduced to 1 month, the cost-effective strategy would become HR-HIVE-1-dose in Côte d'Ivoire and Zimbabwe and HR-HIVE-2-doses in South Africa. Findings regarding the cost-effectiveness of bNAb implementation strategies remained robust in sensitivity analyses regarding breastfeeding duration, maternal engagement in postpartum care, early infant diagnosis uptake and antiretroviral treatment costs. CONCLUSIONS At current efficacy and cost estimates, bNAb prophylaxis for HIV-exposed children in sub-Saharan African settings would be a cost-effective intervention to reduce vertical HIV transmission.
Collapse
Affiliation(s)
- Caitlin M. Dugdale
- Medical Practice Evaluation CenterDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Division of Infectious DiseasesDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Ogochukwu Ufio
- Medical Practice Evaluation CenterDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Christopher Alba
- Medical Practice Evaluation CenterDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Sallie R. Permar
- Department of PediatricsWeill Cornell MedicineNew YorkNew YorkUSA
- Department of PediatricsNew York‐Presbyterian/Weill Cornell Medical CenterNew YorkNew YorkUSA
| | - Lynda Stranix‐Chibanda
- Child and Adolescent Health UnitFaculty of Medicine and Health SciencesUniversity of ZimbabweHarareZimbabwe
| | - Coleen K. Cunningham
- Department of PediatricsUniversity of California IrvineIrvineCaliforniaUSA
- Department of PediatricsChildren's Hospital of Orange CountyOrangeCaliforniaUSA
| | - Genevieve G. Fouda
- Department of PediatricsDuke University Medical CenterDurhamNorth CarolinaUSA
- Duke Human Vaccine InstituteDurhamNorth CarolinaUSA
| | - Landon Myer
- Division of Epidemiology and BiostatisticsSchool of Public Health & Family MedicineUniversity of Cape TownCape TownSouth Africa
| | - Milton C. Weinstein
- Department of Health Policy and ManagementHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Valériane Leroy
- CERPOP, InsermToulouse UniversityUniversité Paul SabatierToulouseFrance
| | - Elizabeth J. McFarland
- Department of PediatricsUniversity of Colorado Anschutz Medical Campus and Children's Hospital ColoradoAuroraColoradoUSA
| | - Kenneth A. Freedberg
- Medical Practice Evaluation CenterDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Division of Infectious DiseasesDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of General Internal MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Andrea L. Ciaranello
- Medical Practice Evaluation CenterDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Division of Infectious DiseasesDepartment of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
17
|
Subtle Longitudinal Alterations in Env Sequence Potentiate Differences in Sensitivity to Broadly Neutralizing Antibodies following Acute HIV-1 Subtype C Infection. J Virol 2022; 96:e0127022. [PMID: 36453881 PMCID: PMC9769376 DOI: 10.1128/jvi.01270-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) for HIV-1 prevention or cure strategies must inhibit transmitted/founder and reservoir viruses. Establishing sensitivity of circulating viruses to bNAbs and genetic patterns affecting neutralization variability may guide rational bNAbs selection for clinical development. We analyzed 326 single env genomes from nine individuals followed longitudinally following acute HIV-1 infection, with samples collected at ~1 week after the first detection of plasma viremia; 300 to 1,709 days postinfection but prior to initiating antiretroviral therapy (ART) (median = 724 days); and ~1 year post ART initiation. Sequences were assessed for phylogenetic relatedness, potential N- and O-linked glycosylation, and variable loop lengths (V1 to V5). A total of 43 env amplicons (median = 3 per patient per time point) were cloned into an expression vector and the TZM-bl assay was used to assess the neutralization profiles of 15 bNAbs targeting the CD4 binding site, V1/V2 region, V3 supersite, MPER, gp120/gp41 interface, and fusion peptide. At 1 μg/mL, the neutralization breadths were as follows: VRC07-LS and N6.LS (100%), VRC01 (86%), PGT151 (81%), 10-1074 and PGT121 (80%), and less than 70% for 10E8, 3BNC117, CAP256.VRC26, 4E10, PGDM1400, and N123-VRC34.01. Features associated with low sensitivity to V1/V2 and V3 bNAbs were higher potential glycosylation sites and/or relatively longer V1 and V4 domains, including known "signature" mutations. The study shows significant variability in the breadth and potency of bNAbs against circulating HIV-1 subtype C envelopes. VRC07-LS, N6.LS, VRC01, PGT151, 10-1074, and PGT121 display broad activity against subtype C variants, and major determinants of sensitivity to most bNAbs were within the V1/V4 domains. IMPORTANCE Broadly neutralizing antibodies (bNAbs) have potential clinical utility in HIV-1 prevention and cure strategies. However, bNAbs target diverse epitopes on the HIV-1 envelope and the virus may evolve to evade immune responses. It is therefore important to identify antibodies with broad activity in high prevalence settings, as well as the genetic patterns that may lead to neutralization escape. We investigated 15 bNAbs with diverse biophysical properties that target six epitopes of the HIV-1 Env glycoprotein for their ability to inhibit viruses that initiated infection, viruses circulating in plasma at chronic infection before antiretroviral treatment (ART), or viruses that were archived in the reservoir during ART in subtype C infected individuals in South Africa, a high burden country. We identify the antibodies most likely to be effective for clinical use in this setting and describe mutational patterns associated with neutralization escape from these antibodies.
Collapse
|
18
|
Backes IM, Byrd BK, Slein MD, Patel CD, Taylor SA, Garland CR, MacDonald SW, Balazs AB, Davis SC, Ackerman ME, Leib DA. Maternally transferred mAbs protect neonatal mice from HSV-induced mortality and morbidity. J Exp Med 2022; 219:e20220110. [PMID: 36156707 PMCID: PMC9516843 DOI: 10.1084/jem.20220110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/29/2022] [Accepted: 09/01/2022] [Indexed: 01/11/2023] Open
Abstract
Neonatal herpes simplex virus (nHSV) infections often result in significant mortality and neurological morbidity despite antiviral drug therapy. Maternally transferred herpes simplex virus (HSV)-specific antibodies reduce the risk of clinically overt nHSV, but this observation has not been translationally applied. Using a neonatal mouse model, we tested the hypothesis that passive transfer of HSV-specific human mAbs can prevent mortality and morbidity associated with nHSV. The mAbs were expressed in vivo via vectored immunoprophylaxis or recombinantly. Through these maternally derived routes or through direct administration to pups, diverse mAbs to HSV glycoprotein D protected against neonatal HSV-1 and HSV-2 infection. Using in vivo bioluminescent imaging, both pre- and post-exposure mAb treatment significantly reduced viral load in mouse pups. Together these studies support the notion that HSV-specific mAb-based therapies could prevent or improve HSV infection outcomes in neonates.
Collapse
Affiliation(s)
- Iara M. Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Brook K. Byrd
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Matthew D. Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Chaya D. Patel
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Sean A. Taylor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | | | | | - Scott C. Davis
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| |
Collapse
|
19
|
Caskey M, Kuritzkes DR. Monoclonal Antibodies as Long-Acting Products: What Are We Learning From Human Immunodeficiency Virus (HIV) and Coronavirus Disease 2019 (COVID-19)? Clin Infect Dis 2022; 75:S530-S540. [PMID: 36410387 PMCID: PMC10200322 DOI: 10.1093/cid/ciac751] [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: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Broadly neutralizing antibodies directed against human immunodeficiency virus (HIV) offer promise as long-acting agents for prevention and treatment of HIV. Progress and challenges are discussed. Lessons may be learned from the development of monoclonal antibodies to treat and prevent COVID-19.
Collapse
Affiliation(s)
| | - Daniel R Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Abrams EJ, Capparelli E, Ruel T, Mirochnick M. Potential of Long-Acting Products to Transform the Treatment and Prevention of Human Immunodeficiency Virus (HIV) in Infants, Children, and Adolescents. Clin Infect Dis 2022; 75:S562-S570. [PMID: 36410381 PMCID: PMC10200315 DOI: 10.1093/cid/ciac754] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Long-acting antiretroviral products have the potential to transform human immunodeficiency virus (HIV) prevention and treatment approaches in pediatric populations. Broadly neutralizing antibodies and/or long-acting antiretroviral formulations by injection could dramatically improve provision of HIV prophylaxis and/or early treatment to newborns and infants at risk of HIV infection. Challenges in daily oral antiretroviral administration to toddlers and school age children living with HIV may be relieved by use of long-acting formulations, but the pharmacokinetics and safety of these products in children must be studied before they can enter routine clinical use. Although some initial studies of broadly neutralizing antibodies and injectable long-acting agents in infants and young children are underway, more studies of these and other long-acting products are needed. For many adolescents, compliance with daily medication administration is especially challenging. Long-acting products hold particular promise for adolescents living with HIV as well as those at high risk of HIV acquisition, and adolescents can usually be included in the drug development pipeline simultaneously with adults. Long-acting products have the potential to provide alternatives to lifelong daily oral drug administration across the pediatric age spectrum, leading to more effective prevention and treatment of HIV infection in infants, children, and adolescents.
Collapse
Affiliation(s)
- Elaine J Abrams
- ICAP at Columbia University, Mailman School of Public Health, Columbia University, New York, New York, USA
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Edmund Capparelli
- Department of Pediatrics and Clinical Pharmacy, University of California San Diego, La Jolla, California, USA
| | - Theodore Ruel
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Mark Mirochnick
- Department of Pediatrics, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Lovelace SE, Helmold Hait S, Yang ES, Fox ML, Liu C, Choe M, Chen X, McCarthy E, Todd JP, Woodward RA, Koup RA, Mascola JR, Pegu A. Anti-viral efficacy of a next-generation CD4-binding site bNAb in SHIV-infected animals in the absence of anti-drug antibody responses. iScience 2022; 25:105067. [PMID: 36157588 PMCID: PMC9490026 DOI: 10.1016/j.isci.2022.105067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/21/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) against HIV-1 are promising immunotherapeutic agents for treatment of HIV-1 infection. bNAbs can be administered to SHIV-infected rhesus macaques to assess their anti-viral efficacy; however, their delivery into macaques often leads to rapid formation of anti-drug antibody (ADA) responses limiting such assessment. Here, we depleted B cells in five SHIV-infected rhesus macaques by pretreatment with a depleting anti-CD20 antibody prior to bNAb infusions to reduce ADA. Peripheral B cells were depleted following anti-CD20 infusions and remained depleted for at least 9 weeks after the 1st anti-CD20 infusion. Plasma viremia dropped by more than 100-fold in viremic animals after the initial bNAb treatment. No significant humoral ADA responses were detected for as long as B cells remained depleted. Our results indicate that transient B cell depletion successfully inhibited emergence of ADA and improved the assessment of anti-viral efficacy of a bNAb in a SHIV-infected rhesus macaque model. Highly potent CD4bs bNAb reduces viremia up to 4 log10 in SHIV-infected animals Sustained B cell depletion prevents development of ADA responses Lack of ADA enables multiple bNAb infusions over 12 weeks
Collapse
Affiliation(s)
- Sarah E Lovelace
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sabrina Helmold Hait
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Madison L Fox
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Cuiping Liu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Misook Choe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xuejun Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Elizabeth McCarthy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - John-Paul Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Ruth A Woodward
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Capparelli EV, Ajibola G, Maswabi K, Holme MP, Bennett K, Powis KM, Moyo S, Mohammed T, Maphorisa C, Hughes MD, Seaton KE, Tomaras GD, Mosher S, Taylor A, O'Connell S, Narpala S, Mcdermott A, Caskey M, Gama L, Lockman S, Jean-Philippe P, Makhema J, Kuritzkes DR, Lichterfeld M, Shapiro RL. Safety and Pharmacokinetics of Intravenous 10-1074 and VRC01LS in Young Children. J Acquir Immune Defic Syndr 2022; 91:182-188. [PMID: 36094485 PMCID: PMC10224771 DOI: 10.1097/qai.0000000000003033] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/26/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Broadly neutralizing monoclonal antibodies (bNAbs) suppress HIV-1 RNA and may deplete residual viral reservoirs. We evaluated the safety and pharmacokinetics (PK) of dual intravenous VRC01LS and 10-1074 in very early-treated children with HIV-1 on suppressive antiretroviral treatment (ART). SETTING Botswana. METHODS Children with HIV-1 (median age 3.1 years) on ART from <7 days old were enrolled. In phase A, 6 children received 10-1074 (30 mg/kg at day 0, 28, and 56) and 6 children received VRC01LS (30 mg/kg at day 0, 10 mg/kg at days 28 and 56) by intravenous infusion. In phase B, 6 children received the 2 bNAbs combined (with higher VRC01LS maintenance dose, 15 mg/kg) every 4 weeks for 32 weeks with PK evaluations over 8 weeks. Population PK models were developed to predict steady-state concentrations. RESULTS BNAb infusions were well tolerated. There were no infusion reactions nor any bNAb-related grade 3 or 4 events. The median (range) first dose Cmax and trough (day 28) combined from both phases were 1405 (876-1999) μg/mL and 133 (84-319) μg/mL for 10-1074 and 776 (559-846) μg/mL and 230 (158-294) μg/mL for VRC01LS. No large differences in bNAb clearances were observed when given in combination. The estimated VRC01LS half-life was shorter than in adults. Predicted steady-state troughs [median (90% prediction interval)] were 261 (95-565) and 266 (191-366) μg/mL for 10-1074 and VRC01LS, respectively, when given in combination. CONCLUSIONS 10-1074 and VRC01LS were safe and well-tolerated among children receiving ART. Troughs exceeded minimal targets with every 4-week administration of 10-1074 at 30 mg/kg and VRC01LS at 15 mg/kg.
Collapse
Affiliation(s)
| | | | - Kenneth Maswabi
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | - Molly P Holme
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Kara Bennett
- Bennett Statistical Consulting Inc, Ballston Lake, NY
| | - Kathleen M Powis
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Departments of Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, MA
| | - Sikhulile Moyo
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
| | | | | | - Michael D Hughes
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Kelly E Seaton
- Center for Human Systems Immunology, Duke University School of Medicine, Department of Surgery, Durham, NC
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Duke University School of Medicine, Department of Surgery, Durham, NC
| | - Shad Mosher
- Center for Human Systems Immunology, Duke University School of Medicine, Department of Surgery, Durham, NC
| | | | | | | | | | - Marina Caskey
- Laboratory of Molecular Immunology, Rockefeller University, New York, NY
| | | | - Shahin Lockman
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA
| | - Patrick Jean-Philippe
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Joseph Makhema
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Daniel R Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | | | - Roger L Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
23
|
What babies need: accelerating access to current and novel antiretroviral drugs in neonates through pharmacokinetic studies. Lancet HIV 2022; 9:e649-e657. [PMID: 35863363 DOI: 10.1016/s2352-3018(22)00121-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/06/2022] [Accepted: 04/13/2022] [Indexed: 11/24/2022]
Abstract
Although 23 antiretroviral drugs are approved for use in adults, only six are approved by regulatory authorities for use in term neonates born to women with HIV, with even fewer options for preterm neonates. A major hurdle for approvals is the delay in the generation of pharmacokinetic and safety data for antiretrovirals in neonates. The median time between the year of approval from the US Food and Drug Administration of an antiretroviral agent for adults and the first publication date for pharmacokinetic data in neonates less than 4 weeks old is 8 years (range 2-23 years). In this Viewpoint, we address pharmacokinetic research gaps and priorities for current and novel antiretroviral use in neonates. We also consider the challenges and provide guidance on neonatal clinical pharmacology research on antiretroviral agents with the goal of stimulating research and expediting the availability of safe medications for the prevention and treatment of HIV in this vulnerable population.
Collapse
|
24
|
Capparelli EV. Pediatric AIDS-Therapeutic Successes Built on a Foundation of Pediatric Clinical Pharmacology with Pharmacokinetic-Pharmacodynamic Modeling. J Pediatr Pharmacol Ther 2022; 27:482-489. [PMID: 36042957 PMCID: PMC9400182 DOI: 10.5863/1551-6776-27.6.482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/10/2022] [Indexed: 09/04/2024]
Affiliation(s)
- Edmund V Capparelli
- Departments of Pediatrics and Clinical Pharmacy (EVC), University of California-San Diego School of Medicine and Skaggs School of Pharmacy and Pharmaceutical Science, La Jolla, CA
| |
Collapse
|
25
|
Berendam SJ, Nelson AN, Yagnik B, Goswami R, Styles TM, Neja MA, Phan CT, Dankwa S, Byrd AU, Garrido C, Amara RR, Chahroudi A, Permar SR, Fouda GG. Challenges and Opportunities of Therapies Targeting Early Life Immunity for Pediatric HIV Cure. Front Immunol 2022; 13:885272. [PMID: 35911681 PMCID: PMC9325996 DOI: 10.3389/fimmu.2022.885272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/16/2022] [Indexed: 11/26/2022] Open
Abstract
Early initiation of antiretroviral therapy (ART) significantly improves clinical outcomes and reduces mortality of infants/children living with HIV. However, the ability of infected cells to establish latent viral reservoirs shortly after infection and to persist during long-term ART remains a major barrier to cure. In addition, while early ART treatment of infants living with HIV can limit the size of the virus reservoir, it can also blunt HIV-specific immune responses and does not mediate clearance of latently infected viral reservoirs. Thus, adjunctive immune-based therapies that are geared towards limiting the establishment of the virus reservoir and/or mediating the clearance of persistent reservoirs are of interest for their potential to achieve viral remission in the setting of pediatric HIV. Because of the differences between the early life and adult immune systems, these interventions may need to be tailored to the pediatric settings. Understanding the attributes and specificities of the early life immune milieu that are likely to impact the virus reservoir is important to guide the development of pediatric-specific immune-based interventions towards viral remission and cure. In this review, we compare the immune profiles of pediatric and adult HIV elite controllers, discuss the characteristics of cellular and anatomic HIV reservoirs in pediatric populations, and highlight the potential values of current cure strategies using immune-based therapies for long-term viral remission in the absence of ART in children living with HIV.
Collapse
Affiliation(s)
- Stella J. Berendam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States,Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States,*Correspondence: Stella J. Berendam, ; Genevieve G. Fouda,
| | - Ashley N. Nelson
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States,Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Bhrugu Yagnik
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Ria Goswami
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Tiffany M. Styles
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Margaret A. Neja
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Caroline T. Phan
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Sedem Dankwa
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Alliyah U. Byrd
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Carolina Garrido
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Rama R. Amara
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States,Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States,Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States,*Correspondence: Stella J. Berendam, ; Genevieve G. Fouda,
| |
Collapse
|
26
|
Advancing the prevention and treatment of HIV in children: priorities for research and development. THE LANCET HIV 2022; 9:e658-e666. [DOI: 10.1016/s2352-3018(22)00101-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 12/22/2022]
|
27
|
Broadly neutralizing antibodies against HIV-1 and concepts for application. Curr Opin Virol 2022; 54:101211. [DOI: 10.1016/j.coviro.2022.101211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/22/2022] [Accepted: 02/27/2022] [Indexed: 12/21/2022]
|
28
|
Vaccine-Induced, High-Magnitude HIV Env-Specific Antibodies with Fc-Mediated Effector Functions Are Insufficient to Protect Infant Rhesus Macaques against Oral SHIV Infection. mSphere 2022; 7:e0083921. [PMID: 35196125 PMCID: PMC8865927 DOI: 10.1128/msphere.00839-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Improved access to antiretroviral therapy (ART) and antenatal care has significantly reduced in utero and peripartum mother-to-child human immunodeficiency virus (HIV) transmission. However, as breast milk transmission of HIV still occurs at an unacceptable rate, there remains a need to develop an effective vaccine for the pediatric population. Previously, we compared different HIV vaccine strategies, intervals, and adjuvants in infant rhesus macaques to optimize the induction of HIV envelope (Env)-specific antibodies with Fc-mediated effector function. In this study, we tested the efficacy of an optimized vaccine regimen against oral simian-human immunodeficiency virus (SHIV) acquisition in infant macaques. Twelve animals were immunized with 1086.c gp120 protein adjuvanted with 3M-052 in stable emulsion and modified vaccinia Ankara (MVA) virus expressing 1086.c HIV Env. Twelve control animals were immunized with empty MVA. The vaccine prime was given within 10 days of birth, with booster doses being administered at weeks 6 and 12. The vaccine regimen induced Env-specific plasma IgG antibodies capable of antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). Beginning at week 15, infants were exposed orally to escalating doses of heterologous SHIV-1157(QNE)Y173H once a week until infected. Despite the induction of strong Fc-mediated antibody responses, the vaccine regimen did not reduce the risk of infection or time to acquisition compared to controls. However, among vaccinated animals, ADCC postvaccination and postinfection was associated with reduced peak viremia. Thus, nonneutralizing Env-specific antibodies with Fc effector function elicited by this vaccine regimen were insufficient for protection against heterologous oral SHIV infection shortly after the final immunization but may have contributed to control of viremia. IMPORTANCE Women of childbearing age are three times more likely to contract HIV infection than their male counterparts. Poor HIV testing rates coupled with low adherence to antiretroviral therapy (ART) result in a high risk of mother-to-infant HIV transmission, especially during the breastfeeding period. A preventative vaccine could curb pediatric HIV infections, reduce potential health sequalae, and prevent the need for lifelong ART in this population. The results of the current study imply that the HIV Env-specific IgG antibodies elicited by this candidate vaccine regimen, despite a high magnitude of Fc-mediated effector function but a lack of neutralizing antibodies and polyfunctional T cell responses, were insufficient to protect infant rhesus macaques against oral virus acquisition.
Collapse
|
29
|
Immunotherapy with Cell-Based Biological Drugs to Cure HIV-1 Infection. Cells 2021; 11:cells11010077. [PMID: 35011639 PMCID: PMC8750418 DOI: 10.3390/cells11010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 11/17/2022] Open
Abstract
Since its discovery 35 years ago, there have been no therapeutic interventions shown to enable full HIV-1 remission. Combined antiretroviral therapy (cART) has achieved the sustained control of HIV-1 replication, however, the life-long treatment does not eradicate long-lived latently infected reservoirs and can result in multiple side effects including the development of multidrug-resistant escape mutants. Antibody-based treatments have emerged as alternative approaches for a HIV-1 cure. Here, we will review clinical advances in coreceptor-targeting antibodies, with respect to anti-CCR5 antibodies in particular, which are currently being generated to target the early stages of infection. Among the Env-specific antibodies widely accepted as relevant in cure strategies, the potential role of those targeting CD4-induced (CD4i) epitopes of the CD4-binding site (CD4bs) in eliminating HIV-1 infected cells has gained increasing interest and will be presented. Together, with approaches targeting the HIV-1 replication cycle, we will discuss the strategies aimed at boosting and modulating specific HIV-1 immune responses, highlighting the harnessing of TLR agonists for their dual role as latency reverting agents (LRAs) and immune-modulatory compounds. The synergistic combinations of different approaches have shown promising results to ultimately enable a HIV-1 cure.
Collapse
|
30
|
Phelps M, Balazs AB. Contribution to HIV Prevention and Treatment by Antibody-Mediated Effector Function and Advances in Broadly Neutralizing Antibody Delivery by Vectored Immunoprophylaxis. Front Immunol 2021; 12:734304. [PMID: 34603314 PMCID: PMC8479175 DOI: 10.3389/fimmu.2021.734304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/24/2021] [Indexed: 01/11/2023] Open
Abstract
HIV-1 broadly neutralizing antibodies (bNAbs) targeting the viral envelope have shown significant promise in both HIV prevention and viral clearance, including pivotal results against sensitive strains in the recent Antibody Mediated Prevention (AMP) trial. Studies of bNAb passive transfer in infected patients have demonstrated transient reduction of viral load at high concentrations that rebounds as bNAb is cleared from circulation. While neutralization is a crucial component of therapeutic efficacy, numerous studies have demonstrated that bNAbs can also mediate effector functions, such as antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and antibody-dependent complement deposition (ADCD). These functions have been shown to contribute towards protection in several models of HIV acquisition and in viral clearance during chronic infection, however the role of target epitope in facilitating these functions, as well as the contribution of individual innate functions in protection and viral clearance remain areas of active investigation. Despite their potential, the transient nature of antibody passive transfer limits the widespread use of bNAbs. To overcome this, we and others have demonstrated vectored antibody delivery capable of yielding long-lasting expression of bNAbs in vivo. Two clinical trials have shown that adeno-associated virus (AAV) delivery of bNAbs is safe and capable of sustained bNAb expression for over 18 months following a single intramuscular administration. Here, we review key concepts of effector functions mediated by bNAbs against HIV infection and the potential for vectored immunoprophylaxis as a means of producing bNAbs in patients.
Collapse
|