1
|
Farinre O, Anaya T, King AC, Endrias K, Hébert AH, Hill AL, Jean S, Wood JS, Ehnert S, Liang S, Laird GM, Mason RD, Roederer M, Safrit JT, Mavigner M, Chahroudi A. SIV Env RhmAbs + N-803 at ART initiation prolongs viral decay without disrupting reservoir establishment in SIV-infected infant macaques. PLoS Pathog 2025; 21:e1012863. [PMID: 39792949 PMCID: PMC11756789 DOI: 10.1371/journal.ppat.1012863] [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: 10/02/2024] [Revised: 01/23/2025] [Accepted: 12/26/2024] [Indexed: 01/12/2025] Open
Abstract
The latent viral reservoir remains the major barrier to HIV cure, placing the burden of strict adherence to antiretroviral therapy (ART) on people living with HIV to prevent recrudescence of viremia. For infants with perinatally acquired HIV, adherence is anticipated to be a lifelong need. In this study, we tested the hypothesis that administration of ART and viral Envelope-specific rhesus-derived IgG1 monoclonal antibodies (RhmAbs) with or without the IL-15 superagonist N-803 early in infection would limit viral reservoir establishment in SIV-infected infant rhesus macaques. Following initiation of ART at 1-2 weeks after oral SIVmac251 infection, we observed biphasic decay of viremia, with first phase decay significantly faster in the ART + SIV RhmAbs-treated group compared to controls that received only ART. In contrast, the addition of N-803 to ART + SIV RhmAbs significantly slowed both the first and second phase viral decay compared to the ART only group. Treatment with a single dose of N-803 resulted in increased frequency of Ki67 expressing NK, CD8+, and CD4+ T cells. Levels of intact SIV proviruses in CD4+ T cells from blood, lymph nodes, and rectum at week 48 of ART did not differ across groups. Similarly, the time to viral rebound following ART interruption was not impacted by the experimental treatments. These results support the concept that the rebound-competent viral reservoir is formed within days after infection and that targeting only productively infected cells for clearance near the time of ART initiation, even during acute infection, may be insufficient to limit reservoir establishment.
Collapse
Affiliation(s)
- Omotayo Farinre
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Tzoalli Anaya
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Alexis C. King
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kedan Endrias
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Anne H. Hébert
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alison L. Hill
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sherrie Jean
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Jennifer S. Wood
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Stephanie Ehnert
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Shan Liang
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
| | - Gregory M. Laird
- Accelevir Diagnostics, Baltimore, Maryland, United States of America
| | - Rosemarie D. Mason
- Vaccine Research Center, National Institutes of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Mario Roederer
- ImmunoTechnology Section, National Institutes of Allergy and Infectious Diseases, Bethesda, Massachusetts, United States of America
| | | | - Maud Mavigner
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, United States of America
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
2
|
Vijayan K. K. V, De Paris K. Nonhuman primate models of pediatric viral diseases. Front Cell Infect Microbiol 2024; 14:1493885. [PMID: 39691699 PMCID: PMC11649651 DOI: 10.3389/fcimb.2024.1493885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious diseases are the leading cause of death in infants and children under 5 years of age. In utero exposure to viruses can lead to spontaneous abortion, preterm birth, congenital abnormalities or other developmental defects, often resulting in lifelong health sequalae. The underlying biological mechanisms are difficult to study in humans due to ethical concerns and limited sample access. Nonhuman primates (NHP) are closely related to humans, and pregnancy and immune ontogeny in infants are very similar to humans. Therefore, NHP are a highly relevant model for understanding fetal and postnatal virus-host interactions and to define immune mechanisms associated with increased morbidity and mortality in infants. We will discuss NHP models of viruses causing congenital infections, respiratory diseases in early life, and HIV. Cytomegalovirus (CMV) remains the most common cause of congenital defects worldwide. Measles is a vaccine-preventable disease, yet measles cases are resurging. Zika is an example of an emerging arbovirus with devastating consequences for the developing fetus and the surviving infant. Among the respiratory viruses, we will discuss influenza and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We will finish with HIV as an example of a lifelong infection without a cure or vaccine. The review will highlight (i) the impact of viral infections on fetal and infant immune development, (ii) how differences in infant and adult immune responses to infection alter disease outcome, and emphasize the invaluable contribution of pediatric NHP infection models to the design of effective treatment and prevention strategies, including vaccines, for human infants.
Collapse
Affiliation(s)
- Vidya Vijayan K. K.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Center for AIDS Research, University of North Carolina, Chapel Hill, NC, United States
- Children’s Research Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
3
|
Dankwa S, Kosman C, Dennis M, Giorgi EE, Vuong K, Pahountis I, Garza A, Binuya C, McCarthy J, Mayer BT, Ngo JT, Enemuo CA, Carnathan DG, Stanfield-Oakley S, Berendam SJ, Weinbaum C, Engelman K, Magnani DM, Chan C, Ferrari G, Silvestri G, Amara RR, Chahroudi A, Permar SR, Fouda GG, Goswami R. A novel HIV triple broadly neutralizing antibody (bNAb) combination-based passive immunization of infant rhesus macaques achieves durable protective plasma neutralization levels and mediates anti-viral effector functions. PLoS One 2024; 19:e0312411. [PMID: 39527587 PMCID: PMC11554116 DOI: 10.1371/journal.pone.0312411] [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: 11/03/2023] [Accepted: 10/05/2024] [Indexed: 11/16/2024] Open
Abstract
To eliminate vertical HIV transmission and achieve therapy-free viral suppression among children living with HIV, novel strategies beyond antiretroviral therapy (ART) are necessary. Our group previously identified a triple broadly neutralizing antibody (bNAb) combination comprising of 3BNC117, PGDM1400 and PGT151 that mediates robust in vitro neutralization and non-neutralizing effector functions against a cross-clade panel of simian human immunodeficiency viruses (SHIVs). In this study, we evaluated the safety, pharmacokinetics, and antiviral potency of this bNAb combination in infant rhesus macaques (RMs). We demonstrate that subcutaneous infusion of the triple bNAb regimen was well tolerated in pediatric monkeys and resulted in durable systemic and mucosal distribution. Plasma obtained from passively-immunized RMs demonstrated potent HIV-neutralizing and Fc-mediated antiviral effector functions. Finally, using the predicted serum neutralization 80% inhibitory dilution titer (PT80) biomarker threshold of >200, which was recently identified as a surrogate endpoint for evaluation of the preventative efficacy of bNAbs against mucosal viral acquisition in human clinical trials, we demonstrated that our regimen has PT80>200 against a large panel of plasma and breast milk-derived HIV strains and cross-clade SHIV variants. This data will guide the development of combination bNAbs for eliminating vertical HIV transmission and for achieving ART-free viral suppression among children living with HIV.
Collapse
Affiliation(s)
- Sedem Dankwa
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
| | - Christina Kosman
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
| | - Maria Dennis
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
| | - Elena E. Giorgi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Kenneth Vuong
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
| | - Ioanna Pahountis
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
| | - Ashley Garza
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States of America
| | - Christian Binuya
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
| | - Janice McCarthy
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, United States of America
- Center for Human Systems Immunology, Duke University, Durham, NC, United States of America
| | - Bryan T. Mayer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Julia T. Ngo
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Chiamaka A. Enemuo
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Diane G. Carnathan
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America
| | - Sherry Stanfield-Oakley
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States of America
| | - Stella J. Berendam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States of America
| | - Carolyn Weinbaum
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
| | - Kathleen Engelman
- Department of Medicine, UMass Chan Medical School, Worcester, MA, United States of America
| | - Diogo M. Magnani
- Department of Medicine, UMass Chan Medical School, Worcester, MA, United States of America
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, United States of America
- Center for Human Systems Immunology, Duke University, Durham, NC, United States of America
| | - Guido Ferrari
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States of America
| | - Guido Silvestri
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Rama R. Amara
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States of America
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
- Gale and Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, New York, NY, United States of America
| | - Genevieve G. Fouda
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
- Gale and Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, New York, NY, United States of America
| | - Ria Goswami
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States of America
- Gale and Ira Drukier Institute for Children’s Health, Weill Cornell Medicine, New York, NY, United States of America
| |
Collapse
|
4
|
Allan‐Blitz L, Mayer KH. Updated guidelines on HIV post-exposure prophylaxis: continued efforts towards increased accessibility. J Int AIDS Soc 2024; 27:e26393. [PMID: 39576221 PMCID: PMC11583823 DOI: 10.1002/jia2.26393] [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: 08/05/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
INTRODUCTION HIV transmission is ongoing in both high- and low-resource settings. Post-exposure prophylaxis (PEP) remains an important tool in preventing HIV; however, PEP is significantly underutilized. The multitude of barriers to PEP implementation include low patient and provider awareness and acceptability, limited access to treatment and prevention services, and high rates of stigma. The World Health Organization (WHO) recently released updated guidance on the delivery of HIV PEP. This commentary aims to highlight the salient changes, evaluate how such recommendations can overcome the existing barriers to PEP implementation and discuss strategies needed to put the updated guidance into practice. DISCUSSION The 2024 WHO PEP guidelines continue a trend towards increasing access to PEP. Most notably, the WHO now provides strong recommendations that: (1) PEP be delivered in community settings (e.g. pharmacies, police stations and online platforms), and (2) PEP delivery and monitoring be done via task sharing involving non-specialist health workers (e.g. pharmacists or community health workers). The guidelines also emphasize that the PEP encounter is an important educable moment whereby a transition to pre-exposure prophylaxis among individuals at continued risk for HIV infection should be discussed. The decentralization of PEP delivery has the potential to overcome numerous barriers to PEP implementation, reduce time to initiation and support adherence with the 28-day course. To translate the recommendations into delivery programmes, however, much more work is needed. Detailed templates can help overcome the heterogeneity of both the community settings in which PEP can now be provided and the populations (e.g. survivors of sexual assault, healthcare workers, sex workers, etc.) among whom PEP may be indicated. Training of the workforce will be essential, which should include, as emphasized by the WHO, training in trauma-based care. Novel formulations of and delivery mechanisms for PEP are also emerging, and how such iterations can synergize with decentralized PEP delivery programmes remains to be seen. CONCLUSIONS The updated WHO PEP guidelines make major strides towards increasing access to PEP. Realization of such aims will require ongoing evaluation and support given the heterogeneity in who benefits most from PEP.
Collapse
Affiliation(s)
- Lao‐Tzu Allan‐Blitz
- Division of Global Health Equity, Department of MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Kenneth H. Mayer
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
- The Fenway Institute of Fenway HealthBostonMassachusettsUSA
- Fenway HealthBostonMassachusettsUSA
- Division of Infectious Diseases, Department of MedicineBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| |
Collapse
|
5
|
Dias J, Fabozzi G, Fourati S, Chen X, Liu C, Ambrozak DR, Ransier A, Laboune F, Hu J, Shi W, March K, Maximova AA, Schmidt SD, Samsel J, Talana CA, Ernste K, Ko SH, Lucas ME, Radecki PE, Boswell KL, Nishimura Y, Todd JP, Martin MA, Petrovas C, Boritz EA, Doria-Rose NA, Douek DC, Sékaly RP, Lifson JD, Asokan M, Gama L, Mascola JR, Pegu A, Koup RA. Administration of anti-HIV-1 broadly neutralizing monoclonal antibodies with increased affinity to Fcγ receptors during acute SHIV AD8-EO infection. Nat Commun 2024; 15:7461. [PMID: 39198422 PMCID: PMC11358508 DOI: 10.1038/s41467-024-51848-y] [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: 03/05/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Anti-HIV-1 broadly neutralizing antibodies (bNAbs) have the dual potential of mediating virus neutralization and antiviral effector functions through their Fab and Fc domains, respectively. So far, bNAbs with enhanced Fc effector functions in vitro have only been tested in NHPs during chronic simian-HIV (SHIV) infection. Here, we investigate the effects of administering in acute SHIVAD8-EO infection either wild-type (WT) bNAbs or bNAbs carrying the S239D/I332E/A330L (DEL) mutation, which increases binding to FcγRs. Emergence of virus in plasma and lymph nodes (LNs) was delayed by bNAb treatment and occurred earlier in monkeys given DEL bNAbs than in those given WT bNAbs, consistent with faster clearance of DEL bNAbs from plasma. DEL bNAb-treated monkeys had higher levels of circulating virus-specific IFNγ single-producing CD8+ CD69+ T cells than the other groups. In LNs, WT bNAbs were evenly distributed between follicular and extrafollicular areas, but DEL bNAbs predominated in the latter. At week 8 post-challenge, LN monocytes and NK cells from DEL bNAb-treated monkeys upregulated proinflammatory signaling pathways and LN T cells downregulated TNF signaling via NF-κB. Overall, bNAbs with increased affinity to FcγRs shape innate and adaptive cellular immunity, which may be important to consider in future strategies of passive bNAb therapy.
Collapse
Affiliation(s)
- Joana Dias
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Giulia Fabozzi
- Tissue Analysis Core, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Slim Fourati
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Xuejun Chen
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cuiping Liu
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David R Ambrozak
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amy Ransier
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Farida Laboune
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jianfei Hu
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wei Shi
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kylie March
- Tissue Analysis Core, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anna A Maximova
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephen D Schmidt
- Humoral Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jakob Samsel
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Institute for Biomedical Sciences, George Washington University, Washington, D.C., USA
| | - Chloe A Talana
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Keenan Ernste
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sung Hee Ko
- Virus Persistence and Dynamics Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Margaret E Lucas
- Virus Persistence and Dynamics Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pierce E Radecki
- Virus Persistence and Dynamics Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kristin L Boswell
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yoshiaki Nishimura
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John-Paul Todd
- Translational Research Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Malcolm A Martin
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eli A Boritz
- Virus Persistence and Dynamics Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicole A Doria-Rose
- Humoral Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rafick-Pierre Sékaly
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Mangaiarkarasi Asokan
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lucio Gama
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Amarendra Pegu
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Shapiro MB, Ordonez T, Pandey S, Mahyari E, Onwuzu K, Reed J, Sidener H, Smedley J, Colgin LM, Johnson A, Lewis AD, Bimber B, Sacha JB, Hessell AJ, Haigwood NL. Immune perturbation following SHIV infection is greater in newborn macaques than in infants. JCI Insight 2024; 9:e144448. [PMID: 39190496 PMCID: PMC11466190 DOI: 10.1172/jci.insight.144448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Transmission of HIV-1 to newborns and infants remains high, with 130,000 new infections in 2022 in resource-limited settings. Half of HIV-infected newborns, if untreated, progress to disease and death within 2 years. While immunologic immaturity likely promotes pathogenesis and poor viral control, little is known about immune damage in newborns and infants. Here we examined pathologic, virologic, and immunologic outcomes in rhesus macaques exposed to pathogenic simian-human immunodeficiency virus (SHIV) at 1-2 weeks, defined as newborns, or at 4 months of age, considered infants. Kinetics of plasma viremia and lymph node seeding DNA were indistinguishable in newborns and infants, but levels of viral DNA in gut and lymphoid tissues 6-10 weeks after infection were significantly higher in newborns versus either infant or adult macaques. Two of 6 newborns with the highest viral seeding required euthanasia at 25 days. We observed age-dependent alterations in leukocyte subsets and gene expression. Compared with infants, newborns had stronger skewing of monocytes and CD8+ T cells toward differentiated subsets and little evidence of type I interferon responses by transcriptomic analyses. Thus, SHIV infection reveals distinct immunological alterations in newborn and infant macaques. These studies lay the groundwork for understanding how immune maturation affects pathogenesis in pediatric HIV-1 infection.
Collapse
Affiliation(s)
- Mariya B. Shapiro
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | - Eisa Mahyari
- Division of Pathobiology & Immunology and
- Genetics Division, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Kosiso Onwuzu
- Division of Pathobiology & Immunology and
- Genetics Division, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jason Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Heather Sidener
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Lois M. Colgin
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Amanda Johnson
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Anne D. Lewis
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Benjamin Bimber
- Division of Pathobiology & Immunology and
- Genetics Division, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jonah B. Sacha
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
- Division of Pathobiology & Immunology and
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Nancy L. Haigwood
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
- Division of Pathobiology & Immunology and
| |
Collapse
|
7
|
Pitchai FNN, Tanner EJ, Khetan N, Vasen G, Levrel C, Kumar AJ, Pandey S, Ordonez T, Barnette P, Spencer D, Jung SY, Glazier J, Thompson C, Harvey-Vera A, Son HI, Strathdee SA, Holguin L, Urak R, Burnett J, Burgess W, Busman-Sahay K, Estes JD, Hessell A, Fennessey CM, Keele BF, Haigwood NL, Weinberger LS. Engineered deletions of HIV replicate conditionally to reduce disease in nonhuman primates. Science 2024; 385:eadn5866. [PMID: 39116226 PMCID: PMC11545966 DOI: 10.1126/science.adn5866] [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: 12/17/2023] [Accepted: 06/06/2024] [Indexed: 08/10/2024]
Abstract
Antiviral therapies with reduced frequencies of administration and high barriers to resistance remain a major goal. For HIV, theories have proposed that viral-deletion variants, which conditionally replicate with a basic reproductive ratio [R0] > 1 (termed "therapeutic interfering particles" or "TIPs"), could parasitize wild-type virus to constitute single-administration, escape-resistant antiviral therapies. We report the engineering of a TIP that, in rhesus macaques, reduces viremia of a highly pathogenic model of HIV by >3log10 following a single intravenous injection. Animal lifespan was significantly extended, TIPs conditionally replicated and were continually detected for >6 months, and sequencing data showed no evidence of viral escape. A single TIP injection also suppressed virus replication in humanized mice and cells from persons living with HIV. These data provide proof of concept for a potential new class of single-administration antiviral therapies.
Collapse
Affiliation(s)
- Fathima N. Nagoor Pitchai
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology, University of California, San Francisco, CA, USA
| | - Elizabeth J. Tanner
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology, University of California, San Francisco, CA, USA
| | - Neha Khetan
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology, University of California, San Francisco, CA, USA
| | - Gustavo Vasen
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology, University of California, San Francisco, CA, USA
| | - Clara Levrel
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology, University of California, San Francisco, CA, USA
| | - Arjun J. Kumar
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology, University of California, San Francisco, CA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Tracy Ordonez
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - David Spencer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
- Absci Corporation, Vancouver, WA, USA
| | - Seung-Yong Jung
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
| | - Joshua Glazier
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
| | - Cassandra Thompson
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alicia Harvey-Vera
- Global Health Sciences, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- US-Mexico Border Health Commission, Tijuana, Mexico
| | - Hye-In Son
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
| | - Steffanie A. Strathdee
- Global Health Sciences, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Leo Holguin
- Global Health Sciences, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ryan Urak
- Center for Gene Therapy, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - John Burnett
- Center for Gene Therapy, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - William Burgess
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Jacob D. Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
- Faculty of Health, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- School of Health and Biomedical Sciences College of Science, Engineering and Health RMIT University, Melbourne, Australia
| | - Ann Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Christine M. Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Leor S. Weinberger
- Gladstone Center for Cell Circuitry, University of California, San Francisco, CA, USA
- Gladstone Institute of Virology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Chinunga TT, Chahroudi A, Ribeiro SP. Pediatric immunotherapy and HIV control. Curr Opin HIV AIDS 2024; 19:201-211. [PMID: 38841850 PMCID: PMC11155294 DOI: 10.1097/coh.0000000000000857] [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: 06/07/2024]
Abstract
PURPOSE OF REVIEW Highlighting opportunities/potential for immunotherapy by understanding dynamics of HIV control during pediatric HIV infection with and without antiretroviral therapy (ART), as modeled in Simian immunodeficiency virus (SIV) and Simian-human immunodeficiency virus (SHIV)-infected rhesus macaques and observed in clinical trials. This review outlines mode of transmission, pathogenesis of pediatric HIV, unique aspects of the infant immune system, infant macaque models and immunotherapies. RECENT FINDINGS During the earliest stages of perinatal HIV infection, the infant immune system is characterized by a unique environment defined by immune tolerance and lack of HIV-specific T cell responses which contribute to disease progression. Moreover, primary lymphoid organs such as the thymus appear to play a distinct role in HIV pathogenesis in children living with HIV (CLWH). Key components of the immune system determine the degree of viral control, targets for strategies to induce viral control, and the response to immunotherapy. The pursuit of highly potent broadly neutralizing antibodies (bNAbs) and T cell vaccines has revolutionized the approach to HIV cure. Administration of HIV-1-specific bNAbs, targeting the highly variable envelope improves humoral immunity, and T cell vaccines induce or improve T cell responses such as the cytotoxic effects of HIV-1-specific CD8+ T cells, both of which are promising options towards virologic control and ART-free remission as evidenced by completed and ongoing clinical trials. SUMMARY Understanding early events during HIV infection and disease progression in CLWH serves as a foundation for predicting or targeting later outcomes by harnessing the immune system's natural responses. The developing pediatric immune system offers multiple opportunities for specific long-term immunotherapies capable of improving quality of life during adolescence and adulthood.
Collapse
Affiliation(s)
- Tehillah T. Chinunga
- Program in Immunology and Molecular Pathogenesis, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University
| | - Susan P. Ribeiro
- Pathology Advanced Translational Research Unit (PATRU), Department of Pathology and Laboratory Medicine, Emory University School of Medicine
- Emory Vaccine Center
- Winship Cancer Institute of Emory University, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Webb GM, Pessoa CT, McCullen AJ, Hwang JM, Humkey MC, Thormin-Odum R, Kukula KA, Smedley J, Fischer M, Sciurba J, Bochart RM, Shriver-Munsch C, Ndhlovu LC, Sacha JB. Immune restoration by TIGIT blockade is insufficient to control chronic SIV infection. J Virol 2024; 98:e0027324. [PMID: 38775481 PMCID: PMC11237531 DOI: 10.1128/jvi.00273-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/27/2024] [Indexed: 06/12/2024] Open
Abstract
TIGIT is a negative immune checkpoint receptor associated with T cell exhaustion in cancer and HIV. TIGIT upregulation in virus-specific CD8+ T cells and NK cells during HIV/SIV infection results in dysfunctional effector capabilities. In vitro studies targeting TIGIT on CD8+ T cells suggest TIGIT blockade as a viable strategy to restore SIV-specific T cell responses. Here, we extend these studies in vivo using TIGIT blockage in nonhuman primates in an effort to reverse T cell and NK cell exhaustion in the setting of SIV infection. We demonstrate that in vivo administration of a humanized anti-TIGIT monoclonal antibody (mAb) is well tolerated in both cynomolgus macaques and rhesus macaques. Despite sustained plasma concentrations of anti-TIGIT mAb, we observed no consistent improvement in NK or T cell cytolytic capacity. TIGIT blockade minimally enhanced T cell proliferation and virus-specific T cell responses in both magnitude and breadth though plasma viral loads in treated animals remained stable indicating that anti-TIGIT mAb treatment alone was insufficient to increase anti-SIV CD8+ T cell function. The enhancement of virus-specific T cell proliferative responses observed in vitro with single or dual blockade of TIGIT and/or PD-1 highlights TIGIT as a potential target to reverse T cell dysfunction. Our studies, however, reveal that targeting the TIGIT pathway alone may be insufficient in the setting of viremia and that combining immune checkpoint blockade with other immunotherapeutics may be a future path forward for improved viral control or elimination of HIV.IMPORTANCEUpregulation of the immune checkpoint receptor TIGIT is associated with HIV-mediated T cell dysfunction and correlates with HIV disease progression. Compelling evidence exists for targeting immune checkpoint receptor pathways that would potentially enhance immunity and refocus effector cell efforts toward viral clearance. In this report, we investigate TIGIT blockade as an immunotherapeutic approach to reverse immune exhaustion during chronic SIV/SHIV infection in a nonhuman primate model of HIV infection. We show that interfering with the TIGIT signaling axis alone is insufficient to improve viral control despite modest improvement in T cell immunity. Our data substantiate the use of targeting multiple immune checkpoint receptors to promote synergy and ultimately eliminate HIV-infected cells.
Collapse
Affiliation(s)
- Gabriela M. Webb
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Cleiton T. Pessoa
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Allyson J. McCullen
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Joseph M. Hwang
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Matthew C. Humkey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Raymond Thormin-Odum
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Kaitlyn A. Kukula
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Miranda Fischer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Joseph Sciurba
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Rachele M. Bochart
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Christine Shriver-Munsch
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Lishomwa C. Ndhlovu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Jonah B. Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
10
|
Fonseca JA, King AC, Chahroudi A. More than the Infinite Monkey Theorem: NHP Models in the Development of a Pediatric HIV Cure. Curr HIV/AIDS Rep 2024; 21:11-29. [PMID: 38227162 PMCID: PMC10859349 DOI: 10.1007/s11904-023-00686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2023] [Indexed: 01/17/2024]
Abstract
PURPOSE OF REVIEW An HIV cure that eliminates the viral reservoir or provides viral control without antiretroviral therapy (ART) is an urgent need in children as they face unique challenges, including lifelong ART adherence and the deleterious effects of chronic immune activation. This review highlights the importance of nonhuman primate (NHP) models in developing an HIV cure for children as these models recapitulate the viral pathogenesis and persistence. RECENT FINDINGS Several cure approaches have been explored in infant NHPs, although knowledge gaps remain. Broadly neutralizing antibodies (bNAbs) show promise for controlling viremia and delaying viral rebound after ART interruption but face administration challenges. Adeno-associated virus (AAV) vectors hold the potential for sustained bNAb expression. Therapeutic vaccination induces immune responses against simian retroviruses but has yet to impact the viral reservoir. Combining immunotherapies with latency reversal agents (LRAs) that enhance viral antigen expression should be explored. Current and future cure approaches will require adaptation for the pediatric immune system and unique features of virus persistence, for which NHP models are fundamental to assess their efficacy.
Collapse
Affiliation(s)
- Jairo A Fonseca
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexis C King
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
- Emory+Children's Center for Childhood Infections and Vaccines, Atlanta, GA, USA.
| |
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
|
Wang W, Zhao S, Wu Y, Duan W, Li S, Li Z, Guo C, Wang W, Zhang T, Wu H, Huang X. Safety and Efficacy of Long-Acting Injectable Agents for HIV-1: Systematic Review and Meta-Analysis. JMIR Public Health Surveill 2023; 9:e46767. [PMID: 37498645 PMCID: PMC10415942 DOI: 10.2196/46767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/13/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND HIV-1 infection continues to affect global health. Although antiretrovirals can reduce the viral load or prevent HIV-1 infection, current drugs require daily oral use with a high adherence level. Long-acting antiretrovirals (LA-ARVs) significantly improve medication adherence and are essential for HIV-1 prophylaxis and therapy. OBJECTIVE This study aimed to investigate the safety and efficacy of long-acting cabotegravir (CAB-LA) and long-acting rilpivirine (RPV-LA) in the prevention and treatment of HIV-1 infection. METHODS PubMed, Embase, and the Cochrane Library were searched for studies from database inception to November 12, 2022. We included studies that reported efficacy and safety data on LA-ARV intervention in people living with HIV and excluded reviews, animal studies, and articles with missing or duplicate data. Virological suppression was defined as plasma viral load <50 copies/mL 6 months after antiviral therapy initiation. We extracted outcomes for analysis and expressed dichotomous data as risk ratios (RRs) and continuous data as mean differences. Depending on the heterogeneity assessment, a fixed- or random-effects model was used for data synthesis. We performed subgroup analyses of the partial safety and efficacy outcomes of CAB-LA+RPV-LA. The protocol was registered with the Open Science Framework. RESULTS We included 12 trials comprising 10,957 individuals, of which 7 were prevention trials and 5 were treatment trials. CAB-LA and RPV-LA demonstrated safety profiles comparable with those of the placebo in terms of adverse event-related withdrawal. Moreover, the efficacy data showed that CAB-LA had a better effect on HIV-1 prevention than tenofovir disoproxil fumarate-emtricitabine (17/5161, 0.33% vs 75/5129, 1.46%; RR 0.21, 95% CI 0.07-0.61; I2=70%). Although CAB-LA+RPV-LA had more drug-related adverse events (556/681, 81.6% vs 37/598, 6.2%; RR 12.50, 95% CI 3.98-39.23; I2=85%), a mild or moderate injection site reaction was the most common reaction, and its frequency decreased over time. The efficacy of CAB-LA+RPV-LA was comparable with that of daily oral drugs at 48 and 96 weeks (1302/1424, 91.43% vs 915/993, 92.2%; RR 0.99, 95% CI 0.97-1.02; I2=0%), and a high level of virological suppression of 80.9% (186/230) was maintained even after 5 years of LA-ARV use. Similar efficacy outcomes were observed in both treatment-naive and treatment-experienced patients (849/911, 93.2% vs 615/654, 94%; RR 0.99, 95% CI 0.96-1.02; I2=0%). According to the questionnaires, more than 85% of people living with HIV favored LA-ARVs. CONCLUSIONS LA-ARVs showed favorable safety profiles for both the prevention and treatment of HIV-1 infection and were well tolerated. CAB-LA has more satisfactory efficacy than tenofovir disoproxil fumarate-emtricitabine, significantly reducing the rate of HIV-1 infection. CAB-LA+RPV-LA maintains virological suppression for a long time and may be a viable switching strategy with enhanced public health benefits by reducing transmission. However, further trials are required to confirm the efficacy of these drugs.
Collapse
Affiliation(s)
- Wenjing Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Shengnan Zhao
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yaxin Wu
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wenshan Duan
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Sibo Li
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Caiping Guo
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wen Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hao Wu
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiaojie Huang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
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
|
14
|
Rosenberg YJ, Ordonez T, Khanwalkar US, Barnette P, Pandey S, Backes IM, Otero CE, Goldberg BS, Crowley AR, Leib DA, Shapiro MB, Jiang X, Urban LA, Lees J, Hessell AJ, Permar S, Haigwood NL, Ackerman ME. Evidence for the Role of a Second Fc-Binding Receptor in Placental IgG Transfer in Nonhuman Primates. mBio 2023; 14:e0034123. [PMID: 36946726 PMCID: PMC10127586 DOI: 10.1128/mbio.00341-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
Transplacental transfer of maternal antibodies provides the fetus and newborn with passive protection against infectious diseases. While the role of the highly conserved neonatal Fc receptor (FcRn) in transfer of IgG in mammals is undisputed, recent reports have suggested that a second receptor may contribute to transport in humans. We report poor transfer efficiency of plant-expressed recombinant HIV-specific antibodies, including engineered variants with high FcRn affinity, following subcutaneous infusion into rhesus macaques close to parturition. Unexpectedly, unlike those derived from mammalian tissue culture, plant-derived antibodies were essentially unable to cross macaque placentas. This defect was associated with poor Fcγ receptor binding and altered Fc glycans and was not recapitulated in mice. These results suggest that maternal-fetal transfer of IgG across the three-layer primate placenta may require a second receptor and suggest a means of providing maternal antibody treatments during pregnancy while avoiding fetal harm. IMPORTANCE This study compared the ability of several human HIV envelope-directed monoclonal antibodies produced in plants with the same antibodies produced in mammalian cells for their ability to cross monkey and mouse placentas. We found that the two types of antibodies have comparable transfer efficiencies in mice, but they are differentially transferred across macaque placentas, consistent with a two-receptor IgG transport model in primates. Importantly, plant-produced monoclonal antibodies have excellent binding characteristics for human FcRn receptors, permitting desirable pharmacokinetics in humans. The lack of efficient transfer across the primate placenta suggests that therapeutic plant-based antibody treatments against autoimmune diseases and cancer could be provided to the mother while avoiding transfer and preventing harm to the fetus.
Collapse
Affiliation(s)
| | - Tracy Ordonez
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Iara M. Backes
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Claire E. Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | | | - Andrew R. Crowley
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - David A. Leib
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mariya B. Shapiro
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | | | | | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
15
|
Hahn PA, Martins MA. Adeno-associated virus-vectored delivery of HIV biologics: the promise of a "single-shot" functional cure for HIV infection. J Virus Erad 2023; 9:100316. [PMID: 36915910 PMCID: PMC10005911 DOI: 10.1016/j.jve.2023.100316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
The ability of immunoglobulin-based HIV biologics (Ig-HIV), including broadly neutralizing antibodies, to suppress viral replication in pre-clinical and clinical studies illustrates how these molecules can serve as alternatives or adjuncts to antiretroviral therapy for treating HIV infection. However, the current paradigm for delivering Ig-HIVs requires repeated passive infusions, which faces both logistical and economic challenges to broad-scale implementation. One promising way to overcome these obstacles and achieve sustained expression of Ig-HIVs in vivo involves the transfer of Ig-HIV genes to host cells utilizing adeno-associated virus (AAV) vectors. Because AAV vectors are non-pathogenic and their genomes persist in the cell nucleus as episomes, transgene expression can last for as long as the AAV-transduced cell lives. Given the long lifespan of myocytes, skeletal muscle is a preferred tissue for AAV-based immunotherapies aimed at achieving persistent delivery of Ig-HIVs. Consistent with this idea, recent studies suggest that lifelong immunity against HIV can be achieved from a one-time intramuscular dose of AAV/Ig-HIV vectors. However, realizing the promise of this approach faces significant hurdles, including the potential of AAV-delivered Ig-HIVs to induce anti-drug antibodies and the high AAV seroprevalence in the human population. Here we describe how these host immune responses can hinder AAV/Ig-HIV therapies and review current strategies for overcoming these barriers. Given the potential of AAV/Ig-HIV therapy to maintain ART-free virologic suppression and prevent HIV reinfection in people living with HIV, optimizing this strategy should become a greater priority in HIV/AIDS research.
Collapse
Affiliation(s)
- Patricia A. Hahn
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458, USA
- The Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Mauricio A. Martins
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458, 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: 4.0] [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
|
Tettamanti Boshier FA, Reeves DB, Duke ER, Swan DA, Prlic M, Cardozo-Ojeda EF, Schiffer JT. Substantial uneven proliferation of CD4 + T cells during recovery from acute HIV infection is sufficient to explain the observed expanded clones in the HIV reservoir. J Virus Erad 2022; 8:100091. [PMID: 36582473 PMCID: PMC9792356 DOI: 10.1016/j.jve.2022.100091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 09/08/2022] [Accepted: 10/10/2022] [Indexed: 12/03/2022] Open
Abstract
The HIV reservoir is a population of 1-10 million anatomically dispersed, latently infected memory CD4+ T cells in which HIV DNA is quiescently integrated into human chromosomal DNA. When antiretroviral therapy (ART) is stopped and HIV replication initiates in one of these cells, systemic viral spread resumes, rekindling progression to AIDS. Therefore, HIV latency prevents cure. The detection of many populations of identical HIV sequences at unique integration sites implicates CD4+ T cell proliferation as the critical driver of reservoir sustainment after a prolonged period of effective ART. Initial reservoir formation occurs during the first week of primary infection usually before ART is started. While empirical data indicates that both de novo infection and cellular proliferation generate latently infected cells during early untreated infection, it is not known which of these mechanisms is predominant. We developed a mathematical model that recapitulates the profound depletion and brisk recovery of CD4+ T cells, reservoir creation, and viral load trajectory during primary HIV infection. We extended the model to stochastically simulate individual HIV reservoir clones. This model predicts the first detection of HIV infected clones approximately 5 weeks after infection as has recently been shown in vivo and suggests that substantial, uneven proliferation among clones during the recovery from CD4+ lymphopenia is the most plausible explanation for the observed clonal reservoir distribution during the first year of infection.
Collapse
Affiliation(s)
- Florencia A. Tettamanti Boshier
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Daniel B. Reeves
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Elizabeth R. Duke
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - David A. Swan
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Martin Prlic
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
- Department of Global Health, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - E. Fabian Cardozo-Ojeda
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
| | - Joshua T. Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave., Seattle, WA, 98122, USA
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
- Clinical Research Division, University of Washington, 1959 NE Pacific St., Seattle, WA, USA
| |
Collapse
|
18
|
Narayanan E, Falcone S, Elbashir SM, Attarwala H, Hassett K, Seaman MS, Carfi A, Himansu S. Rational Design and In Vivo Characterization of mRNA-Encoded Broadly Neutralizing Antibody Combinations against HIV-1. Antibodies (Basel) 2022; 11:67. [PMID: 36412833 PMCID: PMC9680504 DOI: 10.3390/antib11040067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/30/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022] Open
Abstract
Monoclonal antibodies have been used successfully as recombinant protein therapy; however, for HIV, multiple broadly neutralizing antibodies may be necessary. We used the mRNA-LNP platform for in vivo co-expression of 3 broadly neutralizing antibodies, PGDM1400, PGT121, and N6, directed against the HIV-1 envelope protein. mRNA-encoded HIV-1 antibodies were engineered as single-chain Fc (scFv-Fc) to overcome heavy- and light-chain mismatch. In vitro neutralization breadth and potency of the constructs were compared to their parental IgG form. We assessed the ability of these scFv-Fcs to be expressed individually and in combination in vivo, and neutralization and pharmacokinetics were compared to the corresponding full-length IgGs. Single-chain PGDM1400 and PGT121 exhibited neutralization potency comparable to parental IgG, achieving peak systemic concentrations ≥ 30.81 μg/mL in mice; full-length N6 IgG achieved a peak concentration of 974 μg/mL, but did not tolerate single-chain conversion. The mRNA combination encoding full-length N6 IgG and single-chain PGDM1400 and PGT121 was efficiently expressed in mice, achieving high systemic concentration and desired neutralization potency. Analysis of mice sera demonstrated each antibody contributed towards neutralization of multiple HIV-1 pseudoviruses. Together, these data show that the mRNA-LNP platform provides a promising approach for antibody-based HIV treatment and is well-suited for development of combination therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael S. Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | |
Collapse
|
19
|
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
|
20
|
Wang X, Vincent E, Siddiqui S, Turnbull K, Lu H, Blair R, Wu X, Watkins M, Ziani W, Shao J, Doyle-Meyers LA, Russell-Lodrigue KE, Bohm RP, Veazey RS, Xu H. Early treatment regimens achieve sustained virologic remission in infant macaques infected with SIV at birth. Nat Commun 2022; 13:4823. [PMID: 35973985 PMCID: PMC9381774 DOI: 10.1038/s41467-022-32554-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 08/04/2022] [Indexed: 01/28/2023] Open
Abstract
Early antiretroviral therapy (ART) in HIV-infected infants generally fails to achieve a sustained state of ART-free virologic remission, even after years of treatment. Our studies show that viral reservoir seeding is different in neonatal macaques intravenously exposed to SIV at birth, in contrast to adults. Furthermore, one month of ART including an integrase inhibitor, initiated at day 3, but not day 4 or 5 post infection, efficiently and rapidly suppresses viremia to undetectable levels. Intervention initiated at day 3 post infection and continued for 9 months achieves a sustained virologic remission in 4 of 5 infants. Collectively, an early intervention strategy within a key timeframe and regimen may result in viral remission or successful post-exposure prophylaxis for neonatal SIV infection, which may be clinically relevant for optimizing treatment strategies for HIV-infected or exposed infants.
Collapse
Affiliation(s)
- Xiaolei Wang
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Eunice Vincent
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Summer Siddiqui
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Katherine Turnbull
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Hong Lu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Robert Blair
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Xueling Wu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Meagan Watkins
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Widade Ziani
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Jiasheng Shao
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Lara A Doyle-Meyers
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Kasi E Russell-Lodrigue
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Rudolf P Bohm
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Ronald S Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, 18703 Three Rivers Road, Covington, LA, 70433, USA.
| |
Collapse
|
21
|
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: 1.0] [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]
|
22
|
Moats C, Cook K, Armantrout K, Crank H, Uttke S, Maher K, Bochart RM, Lawrence G, Axthelm MK, Smedley JV. Antimicrobial prophylaxis does not improve post-surgical outcomes in SIV/SHIV-uninfected or SIV/SHIV-infected macaques (Macaca mulatta and Macaca fascicularis) based on a retrospective analysis. PLoS One 2022; 17:e0266616. [PMID: 35442982 PMCID: PMC9020680 DOI: 10.1371/journal.pone.0266616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/23/2022] [Indexed: 12/11/2022] Open
Abstract
Surgical antimicrobial prophylaxis is indicated when performing contaminated surgeries, when specific surgical implants are placed, and for prolonged surgical procedures. Unnecessary prophylactic antibiotics are often utilized for macaque surgeries, despite medical and veterinary guidelines. In this study we compared complication rates in macaques receiving peripheral lymph node (PLN) and laparoscopic biopsies, with and without antimicrobial prophylaxis. A majority of animals were SIV or SHIV infected at the time of surgery, so we also compared post-operative complication rates based on infection status. We found no significant difference in PLN biopsy complication rates for animals that received antimicrobial prophylaxis versus those that did not. Animals who underwent laparoscopic procedures and received prophylactic antibiotics had a higher complication rate than those who did not receive them. Complication rates did not differ significantly for SIV/SHIV infected versus uninfected animals for both laparoscopic biopsy procedures and PLN biopsy procedures. SIV/SHIV infected animals that underwent PLN biopsies had no significant difference in complication rates with and without antimicrobial prophylaxis, and SIV/SHIV infected animals receiving prophylactic antibiotics for laparoscopic biopsies had a higher complication rate than those that did not. This study suggests that perioperative prophylactic antibiotics have no role in the management of SIV/SHIV-infected and uninfected macaques undergoing clean, minimally invasive surgeries. Additionally, we recommend eliminating unnecessary antibiotic use in study animals due to their potential confounding impacts on research models and their potential to promote antimicrobial resistance.
Collapse
Affiliation(s)
- Cassandra Moats
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Kimberly Cook
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Kimberly Armantrout
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Hugh Crank
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Samantha Uttke
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Kelly Maher
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Rachele M. Bochart
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - George Lawrence
- Director’s Office, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Michael K. Axthelm
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Jeremy V. Smedley
- Infectious Disease Resource, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| |
Collapse
|
23
|
Chang XL, Reed JS, Webb GM, Wu HL, Le J, Bateman KB, Greene JM, Pessoa C, Waytashek C, Weber WC, Hwang J, Fischer M, Moats C, Shiel O, Bochart RM, Crank H, Siess D, Giobbi T, Torgerson J, Agnor R, Gao L, Dhody K, Lalezari JP, Bandar IS, Carnate AM, Pang AS, Corley MJ, Kelly S, Pourhassan N, Smedley J, Bimber BN, Hansen SG, Ndhlovu LC, Sacha JB. Suppression of human and simian immunodeficiency virus replication with the CCR5-specific antibody Leronlimab in two species. PLoS Pathog 2022; 18:e1010396. [PMID: 35358290 PMCID: PMC8970399 DOI: 10.1371/journal.ppat.1010396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/25/2022] [Indexed: 12/28/2022] Open
Abstract
The CCR5-specific antibody Leronlimab is being investigated as a novel immunotherapy that can suppress HIV replication with minimal side effects. Here we studied the virological and immunological consequences of Leronlimab in chronically CCR5-tropic HIV-1 infected humans (n = 5) on suppressive antiretroviral therapy (ART) and in ART-naïve acutely CCR5-tropic SHIV infected rhesus macaques (n = 4). All five human participants transitioned from daily combination ART to self-administered weekly subcutaneous (SC) injections of 350 mg or 700 mg Leronlimab and to date all participants have sustained virologic suppression for over seven years. In all participants, Leronlimab fully occupied CCR5 receptors on peripheral blood CD4+ T cells and monocytes. In ART-naïve rhesus macaques acutely infected with CCR5-tropic SHIV, weekly SC injections of 50 mg/kg Leronlimab fully suppressed plasma viremia in half of the macaques. CCR5 receptor occupancy by Leronlimab occurred concomitant with rebound of CD4+ CCR5+ T-cells in peripheral blood, and full CCR5 receptor occupancy was found in multiple anatomical compartments. Our results demonstrate that weekly, self-administered Leronlimab was safe, well-tolerated, and efficacious for long-term virologic suppression and should be included in the arsenal of safe, easily administered, longer-acting antiretroviral treatments for people living with HIV-1. Trial Registration: ClinicalTrials.gov Identifiers: NCT02175680 and NCT02355184.
Collapse
Affiliation(s)
- Xiao L. Chang
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jason S. Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Gabriela M. Webb
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Helen L. Wu
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jimmy Le
- Quest Clinical Research, San Francisco, California, United States of America
| | - Katherine B. Bateman
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Justin M. Greene
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Cleiton Pessoa
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Courtney Waytashek
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Whitney C. Weber
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Joseph Hwang
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Miranda Fischer
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Cassandra Moats
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Oriene Shiel
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Rachele M. Bochart
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Hugh Crank
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Don Siess
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Travis Giobbi
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jeffrey Torgerson
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Rebecca Agnor
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Lina Gao
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Kush Dhody
- Amarex Clinical Research LLC, Germantown, Maryland, United States of America
| | - Jacob P. Lalezari
- Quest Clinical Research, San Francisco, California, United States of America
| | - Ivo Sah Bandar
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, United States of America
| | - Alnor M. Carnate
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, United States of America
| | - Alina S. Pang
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, United States of America
| | - Michael J. Corley
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, United States of America
| | - Scott Kelly
- CytoDyn Inc., Vancouver, Washington, United States of America
| | | | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Benjamin N. Bimber
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Scott G. Hansen
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Lishomwa C. Ndhlovu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, United States of America
| | - Jonah B. Sacha
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
24
|
Lee WS, Reynaldi A, Amarasena T, Davenport MP, Parsons MS, Kent SJ. Anti-Drug Antibodies in Pigtailed Macaques Receiving HIV Broadly Neutralising Antibody PGT121. Front Immunol 2021; 12:749891. [PMID: 34867979 PMCID: PMC8636046 DOI: 10.3389/fimmu.2021.749891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
Broadly neutralising antibodies (bNAbs) may play an important role in future strategies for HIV control. The development of anti-drug antibody (ADA) responses can reduce the efficacy of passively transferred bNAbs but the impact of ADA is imperfectly understood. We previously showed that therapeutic administration of the anti-HIV bNAb PGT121 (either WT or LALA version) controlled viraemia in pigtailed macaques with ongoing SHIV infection. We now report on 23 macaques that had multiple treatments with PGT121. We found that an increasing number of intravenous doses of PGT121 or human IgG1 isotype control antibodies (2-4 doses) results in anti-PGT121 ADA induction and low plasma concentrations of PGT121. ADA was associated with poor or absent suppression of SHIV viremia. Notably, ADA within macaque plasma recognised another human bNAb 10E8 but did not bind to the variable domains of PGT121, suggesting that ADA were primarily directed against the constant regions of the human antibodies. These findings have implications for the development of preclinical studies examining multiple infusions of human bNAbs.
Collapse
Affiliation(s)
- Wen Shi Lee
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Thakshila Amarasena
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Matthew S Parsons
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, United States.,Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Stephen J Kent
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Melbourne Sexual Health Centre and Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Deeks SG, Archin N, Cannon P, Collins S, Jones RB, de Jong MAWP, Lambotte O, Lamplough R, Ndung'u T, Sugarman J, Tiemessen CT, Vandekerckhove L, Lewin SR. Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2021. Nat Med 2021; 27:2085-2098. [PMID: 34848888 DOI: 10.1038/s41591-021-01590-5] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/27/2021] [Indexed: 12/21/2022]
Abstract
Despite the success of antiretroviral therapy (ART) for people living with HIV, lifelong treatment is required and there is no cure. HIV can integrate in the host genome and persist for the life span of the infected cell. These latently infected cells are not recognized as foreign because they are largely transcriptionally silent, but contain replication-competent virus that drives resurgence of the infection once ART is stopped. With a combination of immune activators, neutralizing antibodies, and therapeutic vaccines, some nonhuman primate models have been cured, providing optimism for these approaches now being evaluated in human clinical trials. In vivo delivery of gene-editing tools to either target the virus, boost immunity or protect cells from infection, also holds promise for future HIV cure strategies. In this Review, we discuss advances related to HIV cure in the last 5 years, highlight remaining knowledge gaps and identify priority areas for research for the next 5 years.
Collapse
Affiliation(s)
- Steven G Deeks
- University of California San Francisco, San Fransisco, CA, USA.
| | - Nancie Archin
- UNC HIV Cure Center, Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Paula Cannon
- University of Southern California, Los Angeles, CA, USA
| | | | - R Brad Jones
- Weill Cornell Medicine, Cornell University, New York, NY, USA
| | | | - Olivier Lambotte
- University Paris Saclay, AP-HP, Bicêtre Hospital, UMR1184 INSERM CEA, Le Kremlin Bicêtre, Paris, France
| | | | - Thumbi Ndung'u
- Africa Health Research Institute and University of KwaZulu-Natal, Durban, South Africa
- University College London, London, UK
- Ragon Institute of MGH, MIT and Harvard University, Cambridge, MA, USA
| | - Jeremy Sugarman
- Berman Institute of Bioethics and Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Caroline T Tiemessen
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Sharon R Lewin
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia.
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| |
Collapse
|
26
|
Goga AE, Van de Perre P, Ngandu N, Nagot N, Abrams EJ, Moodley D, King R, Molès JP, Chirinda W, Scarlatti G, Tylleskär T, Sherman GG, Pillay Y, Dabis F, Gray G. Eliminating HIV transmission through breast milk from women taking antiretroviral drugs. BMJ 2021; 374:n1697. [PMID: 34588170 PMCID: PMC8479590 DOI: 10.1136/bmj.n1697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Ameena Goga and colleagues argue that frequent testing of maternal viral load is needed to eliminate HIV transmission through breast milk in low and middle income settings
Collapse
Affiliation(s)
- Ameena E Goga
- South African Medical Research Council, Pretoria and Cape Town, South Africa
- University of Pretoria, Pretoria, South Africa
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang; CHU Montpellier, Montpellier, France
| | - Nobubelo Ngandu
- South African Medical Research Council, Pretoria and Cape Town, South Africa
| | - Nicolas Nagot
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang; CHU Montpellier, Montpellier, France
| | - Elaine J Abrams
- ICAP at Columbia, Mailman School of Public Health, Columbia University, New York, USA
| | - Dhayendre Moodley
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu Natal, Durban, South Africa
- Centre for AIDS Research in South Africa, Durban, South Africa
| | - Rachel King
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang; CHU Montpellier, Montpellier, France
- UCSF, San Francisco, CA, USA
| | - Jean-Pierre Molès
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang; CHU Montpellier, Montpellier, France
| | - Witness Chirinda
- South African Medical Research Council, Pretoria and Cape Town, South Africa
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Gayle G Sherman
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV & STI, National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa
| | | | - François Dabis
- Agence Nationale de Recherche sur le Sida et les Hépatites Virales (ANRS), Paris, France
- Université Bordeaux, ISPED, Centre INSERM U1219-Bordeaux Population Health, Bordeaux, France
| | - Glenda Gray
- South African Medical Research Council, Pretoria and Cape Town, South Africa
| |
Collapse
|
27
|
Dias J, Fabozzi G, March K, Asokan M, Almasri CG, Fintzi J, Promsote W, Nishimura Y, Todd JP, Lifson JD, Martin MA, Gama L, Petrovas C, Pegu A, Mascola JR, Koup RA. Concordance of immunological events between intrarectal and intravenous SHIVAD8-EO infection when assessed by Fiebig-equivalent staging. J Clin Invest 2021; 131:e151632. [PMID: 34623326 PMCID: PMC8409578 DOI: 10.1172/jci151632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022] Open
Abstract
Primary HIV-1 infection can be classified into six Fiebig stages based on virological and serological laboratory testing, whereas simian-HIV (SHIV) infection in nonhuman primates (NHPs) is defined in time post-infection, making it difficult to extrapolate NHP experiments to the clinics. We identified and extensively characterized the Fiebig-equivalent stages in NHPs challenged intrarectally or intravenously with SHIVAD8-EO. During the first month post-challenge, intrarectally challenged monkeys were up to 1 week delayed in progression through stages. However, regardless of the challenge route, stages I-II predominated before, and stages V-VI predominated after, peak viremia. Decrease in lymph node (LN) CD4+ T cell frequency and rise in CD8+ T cells occurred at stage V. LN virus-specific CD8+ T cell responses, dominated by degranulation and TNF, were first detected at stage V and increased at stage VI. A similar late elevation in follicular CXCR5+ CD8+ T cells occurred, consistent with higher plasma CXCL13 levels at these stages. LN SHIVAD8-EO RNA+ cells were present at stage II, but appeared to decline at stage VI when virions accumulated in follicles. Fiebig-equivalent staging of SHIVAD8-EO infection revealed concordance of immunological events between intrarectal and intravenous infection despite different infection progressions, and can inform comparisons of NHP studies with clinical data.
Collapse
Affiliation(s)
- Joana Dias
- Immunology Laboratory, Vaccine Research Center
| | | | - Kylie March
- Tissue Analysis Core, Vaccine Research Center
| | | | | | | | | | | | - John-Paul Todd
- Translational Research Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | | | - Lucio Gama
- Immunology Laboratory, Vaccine Research Center
| | | | | | | | | |
Collapse
|
28
|
Rosenberg YJ, Jiang X, Cheever T, Coulter FJ, Pandey S, Sack M, Mao L, Urban L, Lees J, Fischer M, Smedley J, Sidener H, Stanton J, Haigwood NL. Protection of Newborn Macaques by Plant-Derived HIV Broadly Neutralizing Antibodies: a Model for Passive Immunotherapy during Breastfeeding. J Virol 2021; 95:e0026821. [PMID: 34190597 PMCID: PMC8387040 DOI: 10.1128/jvi.00268-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/10/2021] [Indexed: 12/16/2022] Open
Abstract
Preventing human immunodeficiency virus (HIV) infection in newborns by vertical transmission remains an important unmet medical need in resource-poor areas where antiretroviral therapy (ART) is not available and mothers and infants cannot be treated prepartum or during the breastfeeding period. In the present study, the protective efficacy of the potent HIV-neutralizing antibodies PGT121 and VRC07-523, both produced in plants, were assessed in a multiple-SHIV (simian-human immunodeficiency virus)-challenge breastfeeding macaque model. Newborn macaques received either six weekly subcutaneous injections with PGT121 alone or as a cocktail of PGT121-LS plus VRC07-523-LS injected three times every 2 weeks. Viral challenge with SHIVSF162P3 was twice weekly over 5.5 weeks using 11 exposures. Despite the transient presence of plasma viral RNA either immediately after the first challenge or as single-point blips, the antibodies prevented a productive infection in all babies with no sustained plasma viremia, compared to viral loads ranging from 103 to 5 × 108 virions/ml in four untreated controls. No virus was detected in peripheral blood mononuclear cells (PBMCs), and only 3 of 159 tissue samples were weakly positive in the treated babies. Newborn macaques proved to be immunocompetent, producing transient anti-Env antibodies and anti-drug antibody (ADA), which were maintained in the circulation after passive broadly neutralizing antibody clearance. ADA responses were directed to the IgG1 Fc CH2-CH3 domains, which has not been observed to date in adult monkeys passively treated with PGT121 or VRC01. In addition, high levels of VRC07-523 anti-idiotypic antibodies in the circulation of one newborn was concomitant with the rapid elimination of VRC07. Plant-expressed antibodies show promise as passive immunoprophylaxis in a breastfeeding model in newborns. IMPORTANCE Plant-produced human neutralizing antibody prophylaxis is highly effective in preventing infection in newborn monkeys during repeated oral exposure, modeling virus in breastmilk, and offers advantages in cost of production and safety. These findings raise the possibility that anti-Env antibodies may contribute to the control of viral replication in this newborn model and that the observed immune responsiveness may be driven by the long-lived presence of immune complexes.
Collapse
Affiliation(s)
| | | | - Tracy Cheever
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Felicity J. Coulter
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Lingjun Mao
- PlantVax Corporation, Rockville, Maryland, USA
| | - Lori Urban
- PlantVax Corporation, Rockville, Maryland, USA
| | | | - Miranda Fischer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Heather Sidener
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jeffrey Stanton
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
29
|
Wen J, Cheever T, Wang L, Wu D, Reed J, Mascola J, Chen X, Liu C, Pegu A, Sacha JB, Lu Y, Haigwood NL, Chen ISY. Improved delivery of broadly neutralizing antibodies by nanocapsules suppresses SHIV infection in the CNS of infant rhesus macaques. PLoS Pathog 2021; 17:e1009738. [PMID: 34283885 PMCID: PMC8323878 DOI: 10.1371/journal.ppat.1009738] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/30/2021] [Accepted: 06/22/2021] [Indexed: 12/31/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) directed to HIV-1 have shown promise at suppressing viremia in animal models. However, the use of bNAbs for the central nervous system (CNS) infection is confounded by poor penetration of the blood brain barrier (BBB). Typically, antibody concentrations in the CNS are extremely low; with levels in cerebrospinal fluid (CSF) only 0.1% of blood concentrations. Using a novel nanotechnology platform, which we term nanocapsules, we show effective transportation of the human bNAb PGT121 across the BBB in infant rhesus macaques upon systemic administration up to 1.6% of plasma concentration. We demonstrate that a single dose of PGT121 encased in nanocapsules when delivered at 48h post-infection delays early acute infection with SHIVSF162P3 in infants, with one of four animals demonstrating viral clearance. Importantly, the nanocapsule delivery of PGT121 improves suppression of SHIV infection in the CNS relative to controls. In patients where HIV-1 is fully suppressed by antiretroviral drugs, HIV-1 still persists in reservoirs. If antiretroviral drugs are stopped, the virus will emerge from these reservoirs and re-seeds systemically. The central nervous system (CNS) is proposed to be a tissue compartment that harbors other HIV-1 reservoirs. A key obstacle that constrains the treatment for the CNS infection is the blood–brain barrier (BBB), a highly restrictive barrier separating the circulating blood from the brain and extracellular fluid in the CNS, which impedes ~98% of the small molecule therapeutics and almost all macromolecules including broadly neutralizing antibodies (bNAbs) directed to HIV-1. Our “nanocapsule” strategy is based on a nanotechnology wherein bNAb molecules are encapsulated within nanocapsules of which the surface contains abundant choline and acetylcholine analogues. This design allows the nanocapsules to effectively cross the BBB to deliver bNAbs into the CNS upon systemic administration and show an impact of bNAb on CNS reservoirs in SHIV infected infant macaques.
Collapse
Affiliation(s)
- Jing Wen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), UCLA AIDS Institute, Los Angeles, California, United States of America
| | - Tracy Cheever
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Lan Wang
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), UCLA AIDS Institute, Los Angeles, California, United States of America
| | - Di Wu
- Department of Chemical and Biomolecular Engineering, School of Engineering, UCLA, Los Angeles, California, United States of America
| | - Jason Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - John Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Xuejun Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Cuiping Liu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland, United States of America
| | - Jonah B Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America.,Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, School of Engineering, UCLA, Los Angeles, California, United States of America
| | - Nancy L Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Irvin S Y Chen
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), UCLA AIDS Institute, Los Angeles, California, United States of America
| |
Collapse
|
30
|
Spencer DA, Shapiro MB, Haigwood NL, Hessell AJ. Advancing HIV Broadly Neutralizing Antibodies: From Discovery to the Clinic. Front Public Health 2021; 9:690017. [PMID: 34123998 PMCID: PMC8187619 DOI: 10.3389/fpubh.2021.690017] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
Despite substantial progress in confronting the global HIV-1 epidemic since its inception in the 1980s, better approaches for both treatment and prevention will be necessary to end the epidemic and remain a top public health priority. Antiretroviral therapy (ART) has been effective in extending lives, but at a cost of lifelong adherence to treatment. Broadly neutralizing antibodies (bNAbs) are directed to conserved regions of the HIV-1 envelope glycoprotein trimer (Env) and can block infection if present at the time of viral exposure. The therapeutic application of bNAbs holds great promise, and progress is being made toward their development for widespread clinical use. Compared to the current standard of care of small molecule-based ART, bNAbs offer: (1) reduced toxicity; (2) the advantages of extended half-lives that would bypass daily dosing requirements; and (3) the potential to incorporate a wider immune response through Fc signaling. Recent advances in discovery technology can enable system-wide mining of the immunoglobulin repertoire and will continue to accelerate isolation of next generation potent bNAbs. Passive transfer studies in pre-clinical models and clinical trials have demonstrated the utility of bNAbs in blocking or limiting transmission and achieving viral suppression. These studies have helped to define the window of opportunity for optimal intervention to achieve viral clearance, either using bNAbs alone or in combination with ART. None of these advances with bNAbs would be possible without technological advancements and expanding the cohorts of donor participation. Together these elements fueled the remarkable growth in bNAb development. Here, we review the development of bNAbs as therapies for HIV-1, exploring advances in discovery, insights from animal models and early clinical trials, and innovations to optimize their clinical potential through efforts to extend half-life, maximize the contribution of Fc effector functions, preclude escape through multiepitope targeting, and the potential for sustained delivery.
Collapse
Affiliation(s)
- David A. Spencer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Mariya B. Shapiro
- Molecular Microbiology & Immunology Department, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
- Molecular Microbiology & Immunology Department, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| |
Collapse
|
31
|
Zhan W, Muhuri M, Tai PWL, Gao G. Vectored Immunotherapeutics for Infectious Diseases: Can rAAVs Be The Game Changers for Fighting Transmissible Pathogens? Front Immunol 2021; 12:673699. [PMID: 34046041 PMCID: PMC8144494 DOI: 10.3389/fimmu.2021.673699] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Conventional vaccinations and immunotherapies have encountered major roadblocks in preventing infectious diseases like HIV, influenza, and malaria. These challenges are due to the high genomic variation and immunomodulatory mechanisms inherent to these diseases. Passive transfer of broadly neutralizing antibodies may offer partial protection, but these treatments require repeated dosing. Some recombinant viral vectors, such as those based on lentiviruses and adeno-associated viruses (AAVs), can confer long-term transgene expression in the host after a single dose. Particularly, recombinant (r)AAVs have emerged as favorable vectors, given their high in vivo transduction efficiency, proven clinical efficacy, and low immunogenicity profiles. Hence, rAAVs are being explored to deliver recombinant antibodies to confer immunity against infections or to diminish the severity of disease. When used as a vaccination vector for the delivery of antigens, rAAVs enable de novo synthesis of foreign proteins with the conformation and topology that resemble those of natural pathogens. However, technical hurdles like pre-existing immunity to the rAAV capsid and production of anti-drug antibodies can reduce the efficacy of rAAV-vectored immunotherapies. This review summarizes rAAV-based prophylactic and therapeutic strategies developed against infectious diseases that are currently being tested in pre-clinical and clinical studies. Technical challenges and potential solutions will also be discussed.
Collapse
Affiliation(s)
- Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Phillip W. L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
32
|
McFarland EJ, Cunningham CK, Muresan P, Capparelli EV, Perlowski C, Morgan P, Smith B, Hazra R, Purdue L, Harding PA, Theron G, Mujuru H, Agwu A, Purswani M, Rathore MH, Flach B, Taylor A, Lin BC, McDermott AB, Mascola JR, Graham BS. Safety, Tolerability, and Pharmacokinetics of a Long-Acting Broadly Neutralizing HIV-1 Monoclonal Antibody VRC01LS in HIV-1-Exposed Newborn Infants. J Infect Dis 2021; 224:1916-1924. [PMID: 34009371 DOI: 10.1093/infdis/jiab229] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Perinatal HIV-1 continues to occur due to barriers to effective antiretroviral prevention that might be mitigated by long-acting broadly neutralizing monoclonal antibodies (bNAbs). METHODS Extended half-life bNAb, VRC01LS, was administered subcutaneously (SC) at 80 mg/dose after birth to HIV-1-exposed, non-breastfed (Cohort 1, n=10) and breastfed (Cohort 2, n=11) infants. Cohort 2 received a second dose (100mg) at 12 weeks. All received antiretroviral prophylaxis. VRC01LS levels were compared to VRC01 levels determined in a prior cohort. RESULTS Local reactions (all Grade <2) occurred in 67% and 20% after Dose 1 and Dose 2, respectively. The weight-banded dose (mean 28.8 mg/kg) of VRC01LS administrated SC achieved a mean +SD plasma level of 222.3 + 71.6 mcg/mL by 24 hours and 44.0 + 11.6 mcg/mL at week 12, prior to Dose 2. The pre-established target of > 50 mcg/mL was attained in 95% and 32% at week 8 and 12, respectively. The terminal half-life was 37-41 days. VRC01LS level after one dose was significantly greater (p=<0.002) than after a VRC01 dose (20mg/kg). No infants acquired HIV-1. CONCLUSIONS VRC01LS was well tolerated with pharmacokinetics that support further studies of more potent long-acting bNAbs as adjunct treatment with ARVs to prevent infant HIV-1 transmission.
Collapse
Affiliation(s)
- Elizabeth J McFarland
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Coleen K Cunningham
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, 27710, USA.,Children's Hospital Of Orange County (CHOC), 333 City Blvd West, Suite 800, Orange, CA, 92868, USA
| | - Petronella Muresan
- Statistical and Data Management Center/ Frontier Science Foundation, Brookline, MA, 02446, USA
| | - Edmund V Capparelli
- Departments of Pediatrics and Pharmacy, UC San Diego Schools of Medicine and Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, 92093, USA
| | | | - Patricia Morgan
- FHI 360, Durham, NC, 27701, USA.,Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH Bethesda, MD, 20892, USA
| | - Betsy Smith
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Rohan Hazra
- Maternal and Pediatric Infectious Disease Branch Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Bethesda, MD, 20892, USA
| | - Lynette Purdue
- National Institute of Allergy and Infectious Diseases, Division of AIDS (Contractor), Bethesda, MD, USA
| | - Paul A Harding
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Gerhard Theron
- Department of Pediatrics, Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Hilda Mujuru
- University of Zimbabwe College of Health Sciences - Clinical Trials Research Centre (UZCHS-CTRC), Harare, Zimbabwe
| | - Allison Agwu
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Murli Purswani
- Department of Pediatrics, BronxCare Health Systems, affiliated with Icahn School of Medicine at Mount Sinai, Bronx, NY, 10457, USA
| | - Mobeen H Rathore
- University of Florida Center for HIV/AIDS Research, Education and Service (UF CARES), University of Florida College of Medicine, Jacksonville, FL, USA
| | - Britta Flach
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH Bethesda, MD, 20892, USA
| | - Alison Taylor
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH Bethesda, MD, 20892, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH Bethesda, MD, 20892, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH Bethesda, MD, 20892, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH Bethesda, MD, 20892, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH Bethesda, MD, 20892, USA
| | | |
Collapse
|
33
|
Van de Perre P, Goga A, Ngandu N, Nagot N, Moodley D, King R, Molès JP, Mosqueira B, Chirinda W, Scarlatti G, Tylleskär T, Dabis F, Gray G. Eliminating postnatal HIV transmission in high incidence areas: need for complementary biomedical interventions. Lancet 2021; 397:1316-1324. [PMID: 33812490 DOI: 10.1016/s0140-6736(21)00570-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 06/06/2020] [Accepted: 10/02/2020] [Indexed: 11/24/2022]
Abstract
The rate of mother-to-child transmission (MTCT) of HIV from breastfeeding is increasing relative to other causes of MTCT. Early effective preconception and antenatal antiretroviral therapy (ART) reduces intrauterine and intrapartum MTCT, whereas maternal post-partum HIV acquisition, untreated maternal HIV, and suboptimal postnatal maternal ART adherence increase the risk of MTCT through breastfeeding. Although the absolute number of cases of MTCT acquired through breastfeeding is decreasing, the rate of decrease is less than the decrease in intrauterine and intrapartum MTCT. Unless current strategies are universally applied, they might not be sufficient to eliminate MTCT due to breastfeeding. Urgent action is needed to evaluate and implement additional preventive biomedical strategies in high HIV prevalence and incidence settings to eliminate MTCT from breastfeeding. Preventive strategies include: pre-exposure prophylaxis in breastfeeding women who have an increased risk of acquiring HIV; postnatal reinforcement strategies, such as maternal retesting for HIV, maternal care reinforcement, and prophylaxis in infants exposed to HIV via breastmilk; and active (vaccine) or passive immunoprophylaxis with long-acting broadly neutralising antibodies.
Collapse
Affiliation(s)
- Philippe Van de Perre
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Antilles University, CHU Montpellier, Montpellier, France.
| | - Ameena Goga
- South African Medical Research Council, Cape Town, South Africa; Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| | - Nobubelo Ngandu
- South African Medical Research Council, Cape Town, South Africa
| | - Nicolas Nagot
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Antilles University, CHU Montpellier, Montpellier, France
| | - Dhayendre Moodley
- Centre for AIDS Research in South Africa, and Department of Obstetrics and Gynaecology, School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Rachel King
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Antilles University, CHU Montpellier, Montpellier, France; School of Medicine, University of California, San Francisco, CA, USA
| | - Jean-Pierre Molès
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Antilles University, CHU Montpellier, Montpellier, France
| | - Beatriz Mosqueira
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, University of Montpellier, Etablissement Français du Sang, Antilles University, CHU Montpellier, Montpellier, France
| | | | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | | | - François Dabis
- Agence Nationale de Recherche sur le Sida et les Hépatites Virales (ANRS), Paris, France; Bordeaux Population Health, INSERM U 1219, ISPED, Université de Bordeaux, Bordeaux, France
| | - Glenda Gray
- South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
34
|
Malherbe DC, Vang L, Mendy J, Barnette PT, Spencer DA, Reed J, Kareko BW, Sather DN, Pandey S, Wibmer CK, Robins H, Fuller DH, Park B, Lakhashe SK, Wilson JM, Axthelm MK, Ruprecht RM, Moore PL, Sacha JB, Hessell AJ, Alexander J, Haigwood NL. Modified Adenovirus Prime-Protein Boost Clade C HIV Vaccine Strategy Results in Reduced Viral DNA in Blood and Tissues Following Tier 2 SHIV Challenge. Front Immunol 2021; 11:626464. [PMID: 33658998 PMCID: PMC7917243 DOI: 10.3389/fimmu.2020.626464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Designing immunogens and improving delivery methods eliciting protective immunity is a paramount goal of HIV vaccine development. A comparative vaccine challenge study was performed in rhesus macaques using clade C HIV Envelope (Env) and SIV Gag antigens. One group was vaccinated using co-immunization with DNA Gag and Env expression plasmids cloned from a single timepoint and trimeric Env gp140 glycoprotein from one of these clones (DNA+Protein). The other group was a prime-boost regimen composed of two replicating simian (SAd7) adenovirus-vectored vaccines expressing Gag and one Env clone from the same timepoint as the DNA+Protein group paired with the same Env gp140 trimer (SAd7+Protein). The env genes were isolated from a single pre-peak neutralization timepoint approximately 1 year post infection in CAP257, an individual with a high degree of neutralization breadth. Both DNA+Protein and SAd7+Protein vaccine strategies elicited significant Env-specific T cell responses, lesser Gag-specific responses, and moderate frequencies of Env-specific TFH cells. Both vaccine modalities readily elicited systemic and mucosal Env-specific IgG but not IgA. There was a higher frequency and magnitude of ADCC activity in the SAd7+Protein than the DNA+Protein arm. All macaques developed moderate Tier 1 heterologous neutralizing antibodies, while neutralization of Tier 1B or Tier 2 viruses was sporadic and found primarily in macaques in the SAd7+Protein group. Neither vaccine approach provided significant protection from viral acquisition against repeated titered mucosal challenges with a heterologous Tier 2 clade C SHIV. However, lymphoid and gut tissues collected at necropsy showed that animals in both vaccine groups each had significantly lower copies of viral DNA in individual tissues compared to levels in controls. In the SAd7+Protein-vaccinated macaques, total and peak PBMC viral DNA were significantly lower compared with controls. Taken together, this heterologous Tier 2 SHIV challenge study shows that combination vaccination with SAd7+Protein was superior to combination DNA+Protein in reducing viral seeding in tissues in the absence of protection from infection, thus emphasizing the priming role of replication-competent SAd7 vector. Despite the absence of correlates of protection, because antibody responses were significantly higher in this vaccine group, we hypothesize that vaccine-elicited antibodies contribute to limiting tissue viral seeding.
Collapse
Affiliation(s)
- Delphine C Malherbe
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Lo Vang
- Emergent BioSolutions, San Diego, CA, United States
| | - Jason Mendy
- Emergent BioSolutions, San Diego, CA, United States
| | - Philip T Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - David A Spencer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Jason Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Bettie W Kareko
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - D Noah Sather
- Department of Pediatrics, University of Washington, Seattle, WA, United States.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Constantinos K Wibmer
- Centre for HIV and STIs, National Institute for Communicable Diseases, of the National Health Laboratory Service, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Harlan Robins
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Deborah H Fuller
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Byung Park
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Samir K Lakhashe
- Department of Virology and Immunology, Southwest National Primate Research Center, San Antonio, TX, United States.,Texas Biomedical Research Institute, San Antonio, TX, United States
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael K Axthelm
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Ruth M Ruprecht
- Department of Virology and Immunology, Southwest National Primate Research Center, San Antonio, TX, United States.,Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Penny L Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases, of the National Health Laboratory Service, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Division of Medical Virology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Jonah B Sacha
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States.,Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States.,Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Ann J Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | | | - Nancy L Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States.,Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
35
|
Systematic Assessment of Antiviral Potency, Breadth, and Synergy of Triple Broadly Neutralizing Antibody Combinations against Simian-Human Immunodeficiency Viruses. J Virol 2021; 95:JVI.01667-20. [PMID: 33177194 DOI: 10.1128/jvi.01667-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/23/2020] [Indexed: 01/29/2023] Open
Abstract
Daily burden and clinical toxicities associated with antiretroviral therapy (ART) emphasize the need for alternative strategies to induce long-term human immunodeficiency virus (HIV) remission upon ART cessation. Broadly neutralizing antibodies (bNAbs) can both neutralize free virions and mediate effector functions against infected cells and therefore represent a leading immunotherapeutic approach. To increase potency and breadth, as well as to limit the development of resistant virus strains, it is likely that bNAbs will need to be administered in combination. It is therefore critical to identify bNAb combinations that can achieve robust polyfunctional antiviral activity against a high number of HIV strains. In this study, we systematically assessed the abilities of single bNAbs and triple bNAb combinations to mediate robust polyfunctional antiviral activity against a large panel of cross-clade simian-human immunodeficiency viruses (SHIVs), which are commonly used as tools for validation of therapeutic strategies targeting the HIV envelope in nonhuman primate models. We demonstrate that most bNAbs are capable of mediating both neutralizing and nonneutralizing effector functions against cross-clade SHIVs, although the susceptibility to V3 glycan-specific bNAbs is highly strain dependent. Moreover, we observe a strong correlation between the neutralization potencies and nonneutralizing effector functions of bNAbs against the transmitted/founder SHIV CH505. Finally, we identify several triple bNAb combinations comprising of CD4 binding site-, V2-glycan-, and gp120-gp41 interface-targeting bNAbs that are capable of mediating synergistic polyfunctional antiviral activities against multiple clade A, B, C, and D SHIVs.IMPORTANCE Optimal bNAb immunotherapeutics will need to mediate multiple antiviral functions against a broad range of HIV strains. Our systematic assessment of triple bNAb combinations against SHIVs will identify bNAbs with synergistic, polyfunctional antiviral activity that will inform the selection of candidate bNAbs for optimal combination designs. The identified combinations can be validated in vivo in future passive immunization studies using the SHIV challenge model.
Collapse
|
36
|
Moyo T, Guleid FH, Schomaker M, Williamson C, Dorfman JR. HIV-1 Subtype C Tier 3 Viruses Have Increased Infectivity Compared to Tier 2 Viruses. AIDS Res Hum Retroviruses 2020; 36:1010-1019. [PMID: 32935560 DOI: 10.1089/aid.2020.0124] [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: 11/12/2022] Open
Abstract
A primary concern of an antibody-based HIV-1 therapy is the virus' ability to rapidly escape antibody responses. Therefore, we investigated the relationships between antibody neutralization sensitivity, viral phenotype, and infectivity in 13 subtype C viruses using a HeLa transfectant-based assay. We observed that the seven tier 3 viruses exhibited higher infectivity than the tier 2 viruses, suggesting that higher neutralization resistance did not have a substantial entry cost. There was no relationship between neutralization resistance and susceptibility to entry inhibitors Maraviroc, PSC RANTES, or the fusion inhibitor T20, indicating that neutralization resistance may not alter these inhibitor target sites. By analyzing glycosylation patterns in 82 subtype C viruses, we found that the presence of an N-linked glycan motif at position N413 and its absence at N332 were the most important predictors of neutralization resistance. In a set of 200 subtype C viruses, tier 3 strains were more resistant than tier 2 or 1B viruses to several broadly neutralizing monoclonal antibodies targeting three different epitopes. This suggests that it is unlikely that resistance to antibodies targeting a single epitope drives overall resistance. In the context of an antibody-based intervention, highly resistant viruses with increased infectivity, circulating in the population, could hinder HIV-1 control since entry of tier 3 viruses is not always selected against. Therefore, for any long-term antibody-based intervention to be globally relevant, it must elicit responses that limit the occurrence of resistance.
Collapse
Affiliation(s)
- Thandeka Moyo
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Fatuma H. Guleid
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Michael Schomaker
- Centre for Infectious Disease Epidemiology and Research, University of Cape Town, Cape Town, South Africa
| | - Carolyn Williamson
- Division of Medical Virology and Institute of Infectious Disease and Molecular Medicine, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Jeffrey R. Dorfman
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
37
|
Pediatric HIV: the Potential of Immune Therapeutics to Achieve Viral Remission and Functional Cure. Curr HIV/AIDS Rep 2020; 17:237-248. [PMID: 32356090 DOI: 10.1007/s11904-020-00495-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW In the absence of antiretroviral therapy (ART), more than 50% of perinatally HIV-infected children die by 2 years of age. Early ART from infancy is therefore a global recommendation and significantly improves immune health, child survival, and disease outcome. However, even early treatment does not prevent or eradicate the latent reservoir necessitating life-long ART. Adherence to life-long ART is challenging for children and longstanding ART during chronic HIV infection led to higher risks of non-AIDS co-morbidities and virologic failure in infected children. Thus, HIV-infected children are an important population for consideration for immune-based interventions to achieve ART-free remission and functional cure. This review summarizes how the uniqueness of the early life immune system can be harnessed for the development of ART-free remission and functional cure, which means complete virus control in absence of ART. In addition, recent advances in therapeutics in the HIV cure field and their potential for the treatment of pediatric HIV infections are discussed. RECENT FINDINGS Preclinical studies and clinical trials demonstrated that immune-based interventions target HIV replication, limit size of virus reservoir, maintain virus suppression, and delay time to virus rebound. However, these studies have been performed so far only in carefully selected HIV-infected adults, highlighting the need to evaluate the efficacy of immune-based therapeutics in HIV-infected children and to design interventions tailored to the early life maturing immune system. Immune-based therapeutics alone or in combination with ART should be actively explored as potential strategies to achieve viral remission and functional cure in HIV-infected pediatric populations.
Collapse
|
38
|
Desikan R, Raja R, Dixit NM. Early exposure to broadly neutralizing antibodies may trigger a dynamical switch from progressive disease to lasting control of SHIV infection. PLoS Comput Biol 2020; 16:e1008064. [PMID: 32817614 PMCID: PMC7462315 DOI: 10.1371/journal.pcbi.1008064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 09/01/2020] [Accepted: 06/17/2020] [Indexed: 01/01/2023] Open
Abstract
Antiretroviral therapy (ART) for HIV-1 infection is life-long. Stopping therapy typically leads to the reignition of infection and progressive disease. In a major breakthrough, recent studies have shown that early initiation of ART can lead to sustained post-treatment control of viremia, raising hopes of long-term HIV-1 remission. ART, however, elicits post-treatment control in a small fraction of individuals treated. Strikingly, passive immunization with broadly neutralizing antibodies (bNAbs) of HIV-1 early in infection was found recently to elicit long-term control in a majority of SHIV-infected macaques, suggesting that HIV-1 remission may be more widely achievable. The mechanisms underlying the control elicited by bNAb therapy, however, remain unclear. Untreated infection typically leads to progressive disease. We hypothesized that viremic control represents an alternative but rarely realized outcome of the infection and that early bNAb therapy triggers a dynamical switch to this outcome. To test this hypothesis, we constructed a model of viral dynamics with bNAb therapy and applied it to analyse clinical data. The model fit quantitatively the complex longitudinal viral load data from macaques that achieved lasting control. The model predicted, consistently with our hypothesis, that the underlying system exhibited bistability, indicating two potential outcomes of infection. The first had high viremia, weak cytotoxic effector responses, and high effector exhaustion, marking progressive disease. The second had low viremia, strong effector responses, and low effector exhaustion, indicating lasting viremic control. Further, model predictions suggest that early bNAb therapy elicited lasting control via pleiotropic effects. bNAb therapy lowers viremia, which would also limit immune exhaustion. Simultaneously, it can improve effector stimulation via cross-presentation. Consequently, viremia may resurge post-therapy, but would encounter a primed effector population and eventually get controlled. ART suppresses viremia but does not enhance effector stimulation, explaining its limited ability to elicit post-treatment control relative to bNAb therapy. In a remarkable advance in HIV cure research, a recent study showed that 3 weekly doses of HIV-1 broadly neutralizing antibodies (bNAbs) soon after infection kept viral levels controlled for years in most macaques treated. If translated to humans, this bNAb therapy may elicit a functional cure, or long-term remission, of HIV-1 infection, eliminating the need for life-long antiretroviral therapy (ART). How early bNAb therapy works remains unknown. Here, we elucidate the mechanism using mathematical modeling and analysis of in vivo data. We predict that early bNAb therapy suppresses viremia, which reduces exhaustion of cytotoxic effector cells, and enhances antigen uptake and effector stimulation. Collectively, these effects drive infection to lasting control. Model predictions based on these effects fit in vivo data quantitatively. ART controls viremia but does not improve effector stimulation, explaining its weaker ability to induce lasting control post-treatment. Our findings may help improve strategies for achieving functional cure of HIV-1 infection.
Collapse
Affiliation(s)
- Rajat Desikan
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
- * E-mail: (RD); (NMD)
| | - Rubesh Raja
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
| | - Narendra M. Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, India
- * E-mail: (RD); (NMD)
| |
Collapse
|
39
|
Gardner MR. Promise and Progress of an HIV-1 Cure by Adeno-Associated Virus Vector Delivery of Anti-HIV-1 Biologics. Front Cell Infect Microbiol 2020; 10:176. [PMID: 32391289 PMCID: PMC7190809 DOI: 10.3389/fcimb.2020.00176] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Despite the success of antiretroviral therapy (ART) at suppressing HIV-1 infection, a cure that eradicates all HIV-1-infected cells has been elusive. The latent viral reservoir remains intact in tissue compartments that are not readily targeted by the host immune response that could accelerate the rate of reservoir decline during ART. However, over the past decade, numerous broadly neutralizing antibodies (bNAbs) have been discovered and characterized. These bNAbs have also given rise to engineered antibody-like inhibitors that are just as or more potent than bNAbs themselves. The question remains whether bNAbs and HIV-1 inhibitors will be the effective “kill” to a shock-and-kill approach to eliminate the viral reservoir. Additional research over the past few years has sought to develop recombinant adeno-associated virus (rAAV) vectors to circumvent the need for continual administration of bNAbs and maintain persistent expression in a host. This review discusses the advancements made in using rAAV vectors for the delivery of HIV-1 bNAbs and inhibitors and the future of this technology in HIV-1 cure research. Numerous groups have demonstrated with great efficacy that rAAV vectors can successfully express protective concentrations of bNAbs and HIV-1 inhibitors. Yet, therapeutic concentrations, especially in non-human primate (NHP) models, are not routinely achieved. As new studies have been reported, more challenges have been identified for utilizing rAAV vectors, specifically how the host immune response limits the attainable concentrations of bNAbs and inhibitors. The next few years should provide improvements to rAAV vector delivery that will ultimately show whether they can be used for expressing bNAbs and HIV-1 inhibitors to eliminate the HIV-1 viral reservoir.
Collapse
Affiliation(s)
- Matthew R Gardner
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, United States
| |
Collapse
|