1
|
Grobben M, Bakker M, Schriek AI, Levels LJ, Umotoy JC, Tejjani K, van Breemen MJ, Lin RN, de Taeye SW, Ozorowski G, Kootstra NA, Ward AB, Kent SJ, Hogarth PM, Wines BD, Sanders RW, Chung AW, van Gils MJ. Polyfunctionality and breadth of HIV-1 antibodies are associated with delayed disease progression. PLoS Pathog 2024; 20:e1012739. [PMID: 39661636 PMCID: PMC11634010 DOI: 10.1371/journal.ppat.1012739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/09/2024] [Indexed: 12/13/2024] Open
Abstract
HIV-1 infection leads to chronic disease requiring life-long treatment and therefore alternative therapeutics, a cure and/or a protective vaccine are needed. Antibody-mediated effector functions could have a role in the fight against HIV-1. However, the properties underlying the potential beneficial effects of antibodies during HIV-1 infection are poorly understood. To identify a specific profile of antibody features associated with delayed disease progression, we studied antibody polyfunctionality during untreated HIV-1 infection in the well-documented Amsterdam Cohort Studies. Serum samples were analyzed from untreated individuals with HIV-1 at approximately 6 months (n = 166) and 3 years (n = 382) post-seroconversion (post-SC). A Luminex antibody Fc array was used to profile 15 different Fc features for serum antibodies against 20 different HIV-1 envelope glycoprotein antigens and the resulting data was also compared with data on neutralization breadth. We found that high HIV-1 specific IgG1 levels and low IgG2 and IgG4 levels at 3 years post-SC were associated with delayed disease progression. Moreover, delayed disease progression was associated with a broad and polyfunctional antibody response. Specifically, the capacity to interact with all Fc γ receptors (FcγRs) and C1q, and in particular with FcγRIIa, correlated positively with delayed disease progression. There were strong correlations between antibody Fc features and neutralization breadth and several antibody features that were associated with delayed disease progression were also associated with the development of broad and potent antibody neutralization. In summary, we identified a strong association between broad, polyfunctional antibodies and delayed disease progression. These findings contribute new information for the fight against HIV-1, especially for new antibody-based therapy and cure strategies.
Collapse
Affiliation(s)
- Marloes Grobben
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Margreet Bakker
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Angela I. Schriek
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Liesbeth J.J. Levels
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Jeffrey C. Umotoy
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Khadija Tejjani
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Mariëlle J. van Breemen
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Ryan N. Lin
- The Scripps Research Institute, Department of Structural Biology and Computational Biology, La Jolla, California, United States of America
| | - Steven W. de Taeye
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Gabriel Ozorowski
- The Scripps Research Institute, Department of Structural Biology and Computational Biology, La Jolla, California, United States of America
| | - Neeltje A. Kootstra
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Amsterdam UMC, location University of Amsterdam, Department of Experimental Immunology, Amsterdam, The Netherlands
| | - Andrew B. Ward
- The Scripps Research Institute, Department of Structural Biology and Computational Biology, La Jolla, California, United States of America
| | - Stephen J. Kent
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Department of Microbiology and Immunology, Melbourne, Australia
- Alfred Hospital and Central Clinical School, Monash University, Melbourne Sexual Health Centre and Department of Infectious Diseases, Melbourne, Australia
| | - P. Mark Hogarth
- Burnet Institute, Immune Therapies Group, Melbourne, Australia
- Central Clinical School, Monash University, Department of Immunology, Melbourne, Australia
| | - Bruce D. Wines
- Burnet Institute, Immune Therapies Group, Melbourne, Australia
- Central Clinical School, Monash University, Department of Immunology, Melbourne, Australia
| | - Rogier W. Sanders
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Weill Medical College of Cornell University, Department of Microbiology and Immunology, New York, New York, United States of America
| | - Amy W. Chung
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Department of Microbiology and Immunology, Melbourne, Australia
| | - Marit J. van Gils
- Amsterdam UMC, location University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Yamaguchi K, Shimizu H, Takahashi K, Nagatomo T, Nishimura T, Matsumoto M, Koshizuka T, Mori H, Inoue N, Torikai M. Characterization of epitopes of human monoclonal antibodies against cytomegalovirus glycoprotein B for neutralization and antibody-dependent phagocytosis. Vaccine 2023; 41:4497-4507. [PMID: 37321896 DOI: 10.1016/j.vaccine.2023.06.003] [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: 02/25/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023]
Abstract
As congenital cytomegalovirus (CMV) infections are the leading non-genetic cause of sensorineural hearing loss and significant neurological disabilities in children, the development of CMV vaccines should be given the highest public health priority. Although MF59-adjuvanted glycoprotein B (gB) vaccine (gB/MF59) is safe and immunogenic, its efficacy in terms of protection from natural infection was around 50 % in clinical trials. Although gB/MF59 induced high antibody titers, anti-gB antibodies contributed little to the neutralization of infection. Recent studies have found that non-neutralizing functions, including antibody-dependent phagocytosis of virions and virus-infected cells, are likely to play important roles in pathogenesis and vaccine design. Previously, we isolated human monoclonal antibodies (MAbs) that reacted with the trimeric form of gB ectodomain and found that preferential epitopes for neutralization were present on Domains (Doms) I and II of gB, while there were abundant non-neutralizing antibodies targeting Dom IV. In this study, we analyzed the phagocytosis activities of these MAbs and found the following: 1) MAbs effective for phagocytosis of the virions targeted Doms I and II, 2) the MAbs effective for phagocytosis of the virions and those of virus-infected cells were generally distinct, and 3) the antibody-dependent phagocytosis showed little correlation with neutralizing activities. Taking account of the frequency and levels of neutralization and phagocytosis, incorporation of the epitopes on Doms I and II into developing vaccines is considered desirable for the prevention of viremia.
Collapse
Affiliation(s)
| | | | - Keita Takahashi
- Microbiology & Immunology, Gifu Pharmaceutical University, Japan
| | | | | | | | - Tetsuo Koshizuka
- Microbiology & Immunology, Gifu Pharmaceutical University, Japan
| | - Hiroaki Mori
- Kikuchi Research Center, KM Biologics Co., Ltd, Japan
| | - Naoki Inoue
- Microbiology & Immunology, Gifu Pharmaceutical University, Japan.
| | | |
Collapse
|
3
|
Anderko RR, Mailliard RB. Mapping the interplay between NK cells and HIV: therapeutic implications. J Leukoc Biol 2023; 113:109-138. [PMID: 36822173 PMCID: PMC10043732 DOI: 10.1093/jleuko/qiac007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Indexed: 01/18/2023] Open
Abstract
Although highly effective at durably suppressing plasma HIV-1 viremia, combination antiretroviral therapy (ART) treatment regimens do not eradicate the virus, which persists in long-lived CD4+ T cells. This latent viral reservoir serves as a source of plasma viral rebound following treatment interruption, thus requiring lifelong adherence to ART. Additionally, challenges remain related not only to access to therapy but also to a higher prevalence of comorbidities with an inflammatory etiology in treated HIV-1+ individuals, underscoring the need to explore therapeutic alternatives that achieve sustained virologic remission in the absence of ART. Natural killer (NK) cells are uniquely positioned to positively impact antiviral immunity, in part due to the pleiotropic nature of their effector functions, including the acquisition of memory-like features, and, therefore, hold great promise for transforming HIV-1 therapeutic modalities. In addition to defining the ability of NK cells to contribute to HIV-1 control, this review provides a basic immunologic understanding of the impact of HIV-1 infection and ART on the phenotypic and functional character of NK cells. We further delineate the qualities of "memory" NK cell populations, as well as the impact of HCMV on their induction and subsequent expansion in HIV-1 infection. We conclude by highlighting promising avenues for optimizing NK cell responses to improve HIV-1 control and effect a functional cure, including blockade of inhibitory NK receptors, TLR agonists to promote latency reversal and NK cell activation, CAR NK cells, BiKEs/TriKEs, and the role of HIV-1-specific bNAbs in NK cell-mediated ADCC activity against HIV-1-infected cells.
Collapse
Affiliation(s)
- Renee R. Anderko
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Robbie B. Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| |
Collapse
|
4
|
Cottignies-Calamarte A, Tudor D, Bomsel M. Antibody Fc-chimerism and effector functions: When IgG takes advantage of IgA. Front Immunol 2023; 14:1037033. [PMID: 36817447 PMCID: PMC9933243 DOI: 10.3389/fimmu.2023.1037033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Recent advances in the development of therapeutic antibodies (Abs) have greatly improved the treatment of otherwise drug-resistant cancers and autoimmune diseases. Antibody activities are mediated by both their Fab and the Fc. However, therapeutic Abs base their protective mechanisms on Fc-mediated effector functions resulting in the activation of innate immune cells by FcRs. Therefore, Fc-bioengineering has been widely used to maximise the efficacy and convenience of therapeutic antibodies. Today, IgG remains the only commercially available therapeutic Abs, at the expense of other isotypes. Indeed, production, sampling, analysis and related in vivo studies are easier to perform with IgG than with IgA due to well-developed tools. However, interest in IgA is growing, despite a shorter serum half-life and a more difficult sampling and purification methods than IgG. Indeed, the paradigm that the effector functions of IgG surpass those of IgA has been experimentally challenged. Firstly, IgA has been shown to bind to its Fc receptor (FcR) on effector cells of innate immunity with greater efficiency than IgG, resulting in more robust IgA-mediated effector functions in vitro and better survival of treated animals. In addition, the two isotypes have been shown to act synergistically. From these results, new therapeutic formats of Abs are currently emerging, in particular chimeric Abs containing two tandemly expressed Fc, one from IgG (Fcγ) and one from IgA (Fcα). By binding both FcγR and FcαR on effector cells, these new chimeras showed improved effector functions in vitro that were translated in vivo. Furthermore, these chimeras retain an IgG-like half-life in the blood, which could improve Ab-based therapies, including in AIDS. This review provides the rationale, based on the biology of IgA and IgG, for the development of Fcγ and Fcα chimeras as therapeutic Abs, offering promising opportunities for HIV-1 infected patients. We will first describe the main features of the IgA- and IgG-specific Fc-mediated signalling pathways and their respective functional differences. We will then summarise the very promising results on Fcγ and Fcα containing chimeras in cancer treatment. Finally, we will discuss the impact of Fcα-Fcγ chimerism in prevention/treatment strategies against infectious diseases such as HIV-1.
Collapse
Affiliation(s)
- Andréa Cottignies-Calamarte
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, Paris, France.,Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Daniela Tudor
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, Paris, France.,Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, Paris, France.,Université Paris Cité, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Paris, France
| |
Collapse
|
5
|
FCGR3A gene duplication, FcγRIIb-232TT and FcγRIIIb-HNA1a associate with an increased risk of vertical acquisition of HIV-1. PLoS One 2022; 17:e0273933. [PMID: 36084039 PMCID: PMC9462732 DOI: 10.1371/journal.pone.0273933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/17/2022] [Indexed: 11/19/2022] Open
Abstract
Background Some mother-to-child transmission (MTCT) studies suggest that allelic variations of Fc gamma receptors (FcγR) play a role in infant HIV-1 acquisition, but findings are inconsistent. To address the limitations of previous studies, the present study investigates the association between perinatal HIV-1 transmission and FcγR variability in three cohorts of South African infants born to women living with HIV-1. Methods This nested case-control study combines FCGR genotypic data from three perinatal cohorts at two hospitals in Johannesburg, South Africa. Children with perinatally-acquired HIV-1 (cases, n = 395) were compared to HIV-1-exposed uninfected children (controls, n = 312). All study participants were black South Africans and received nevirapine for prevention of MTCT. Functional variants were genotyped using a multiplex ligation-dependent probe amplification assay, and their representation compared between groups using logistic regression analyses. Results FCGR3A gene duplication associated with HIV-1 acquisition (OR = 10.27; 95% CI 2.00–52.65; P = 0.005) as did the FcγRIIb-232TT genotype even after adjusting for FCGR3A copy number and FCGR3B genotype (AOR = 1.72; 95%CI 1.07–2.76; P = 0.024). The association between FcγRIIb-232TT genotype and HIV-1 acquisition was further strengthened (AOR = 2.28; 95%CI 1.11–4.69; P = 0.024) if adjusted separately for FCGR2C c.134-96C>T. Homozygous FcγRIIIb-HNA1a did not significantly associate with HIV-1 acquisition in a univariate model (OR = 1.42; 95%CI 0.94–2.16; P = 0.098) but attained significance after adjustment for FCGR3A copy number and FCGR2B genotype (AOR = 1.55; 95%CI 1.01–2.38; P = 0.044). Both FcγRIIb-232TT (AOR = 1.83; 95%CI 1.13–2.97; P = 0.014) and homozygous FcγRIIIb-HNA1a (AOR = 1.66; 95%CI 1.07–2.57; P = 0.025) retained significance when birthweight and breastfeeding were added to the model. The common FCGR2A and FCGR3A polymorphisms did not associate with HIV-1 acquisition. Conclusions Collectively, our findings suggest that the FcγRIIb-232TT genotype exerts a controlling influence on infant susceptibility to HIV-1 infection. We also show a role for less studied variants–FCGR3A duplication and homozygous HNA1a. These findings provide additional insight into a role for FcγRs in HIV-1 infection in children.
Collapse
|
6
|
Lin LY, Carapito R, Su B, Moog C. Fc receptors and the diversity of antibody responses to HIV infection and vaccination. Genes Immun 2022; 23:149-156. [PMID: 35688931 PMCID: PMC9388370 DOI: 10.1038/s41435-022-00175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022]
Abstract
The development of an effective vaccine against HIV is desperately needed. The successive failures of HIV vaccine efficacy trials in recent decades have shown the difficulty of inducing an appropriate protective immune response to fight HIV. Different correlates of antibody parameters associated with a decreased risk of HIV-1 acquisition have been identified. However, these parameters are difficult to reproduce and improve, possibly because they have an intricate and combined action. Here, we describe the numerous antibody (Ab) functions associated with HIV-1 protection and report the interrelated parameters regulating their complex functions. Indeed, besides neutralizing and Fc-mediated activity, additional factors such as Ab type, concentration and kinetics of induction, and Fc-receptor expression and binding capacity also influence the protective effect conferred by Abs. As these parameters were described to be associated with ethnicity, age and sex, these additional factors must be considered for the development of an effective immune response. Therefore, future vaccine designs need to consider these multifaceted Ab functions together with the demographic attributes of the patient populations.
Collapse
Affiliation(s)
- Li-Yun Lin
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Raphael Carapito
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg, France
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Christiane Moog
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France. .,Vaccine Research Institute (VRI), Créteil, France.
| |
Collapse
|
7
|
Vieira VA, Herbert N, Cromhout G, Adland E, Goulder P. Role of Early Life Cytotoxic T Lymphocyte and Natural Killer Cell Immunity in Paediatric HIV Cure/Remission in the Anti-Retroviral Therapy Era. Front Immunol 2022; 13:886562. [PMID: 35634290 PMCID: PMC9130627 DOI: 10.3389/fimmu.2022.886562] [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: 02/28/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Only three well-characterised cases of functional cure have been described in paediatric HIV infection over the past decade. This underlines the fact that early initiation of combination antiretroviral therapy (cART), whilst minimising the size of the viral reservoir, is insufficient to achieve cure, unless other factors contribute. In this review, we consider these additional factors that may facilitate functional cure in paediatric infection. Among the early life immune activity, these include HIV-specific cytotoxic T-lymphocyte (CTL) and natural killer (NK) cell responses. The former have less potent antiviral efficacy in paediatric compared with adult infection, and indeed, in early life, NK responses have greater impact in suppressing viral replication than CTL. This fact may contribute to a greater potential for functional cure to be achieved in paediatric versus adult infection, since post-treatment control in adults is associated less with highly potent CTL activity, and more with effective antiviral NK cell responses. Nonetheless, antiviral CTL responses can play an increasingly effective role through childhood, especially in individuals expressing then 'protective' HLA-I molecules HLA-B*27/57/58:01/8101. The role of the innate system on preventing infection, in shaping the particular viruses transmitted, and influencing outcome is discussed. The susceptibility of female fetuses to in utero mother-to-child transmission, especially in the setting of recent maternal infection, is a curiosity that also provides clues to mechanisms by which cure may be achieved, since initial findings are that viral rebound is less frequent among males who interrupt cART. The potential of broadly neutralising antibody therapy to facilitate cure in children who have received early cART is discussed. Finally, we draw attention to the impact of the changing face of the paediatric HIV epidemic on cure potential. The effect of cART is not limited to preventing AIDS and reducing the risk of transmission. cART also affects which mothers transmit. No longer are mothers who transmit those who carry genes associated with poor immune control of HIV. In the cART era, a high proportion (>70% in our South African study) of transmitting mothers are those who seroconvert in pregnancy or who for social reasons are diagnosed late in pregnancy. As a result, now, genes associated with poor immune control of HIV are not enriched in mothers who transmit HIV to their child. These changes will likely influence the effectiveness of HLA-associated immune responses and therefore cure potential among children.
Collapse
Affiliation(s)
- Vinicius A. Vieira
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Nicholas Herbert
- Africa Health Research Institute (AHRI), Nelson R Mandela School of Medicine, Durban, South Africa
| | - Gabriela Cromhout
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Emily Adland
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom,Africa Health Research Institute (AHRI), Nelson R Mandela School of Medicine, Durban, South Africa,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa,*Correspondence: Philip Goulder,
| |
Collapse
|
8
|
Thomas AS, Coote C, Moreau Y, Isaac JE, Ewing AC, Kourtis AP, Sagar M. Antibody-dependent cellular cytotoxicity (ADCC) responses along with ADCC susceptibility influence HIV-1 mother to child transmission. JCI Insight 2022; 7:159435. [PMID: 35324477 PMCID: PMC9090239 DOI: 10.1172/jci.insight.159435] [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: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND HIV-1 vaccine efforts are primarily directed towards eliciting neutralizing antibodies (nAbs). However, vaccine trials and mother to child natural history cohort investigations indicate that antibody-dependent cellular cytotoxicity (ADCC), not nAbs, correlate with prevention. The ADCC characteristics associated with lack of HIV-1 acquisition remain unclear. METHODS Here we examine ADCC and nAb properties in pre-transmission plasma from HIV-1 exposed infants and from the corresponding transmitting and non-transmitting mothers' breast milk and plasma. Breadth and potency (BP) is assessed against a panel of heterologous, non-maternal, variants. ADCC and neutralization sensitivity is estimated for the strains present in the infected mothers. RESULTS Infants that eventually acquire HIV-1 and those that remain uninfected have similar pre-transmission ADCC BP. The viruses circulating in the transmitting and the non-transmitting mothers also have similar ADCC susceptibility. Infants with a combination of higher pre-transmission ADCC BP and exposure to more ADCC susceptible strains are less likely to acquire HIV-1. In contrast, higher pre-existing infant neutralization BP and greater maternal virus neutralization sensitivity does not associate with transmission. Infants have higher ADCC BP closer to birth and in the presence of high plasma IgG relative to IgA levels. Mothers with potent humoral responses against their autologous viruses harbor more ADCC sensitive strains. CONCLUSION ADCC sensitivity of the exposure variants along with preexisting ADCC BP influence mother to child HIV-1 transmission during breastfeeding. Vaccination strategies that enhance ADCC responses are likely not sufficient to prevent HIV-1 transmission because strains present in chronically infected individuals can have low ADCC susceptibility. TRIAL REGISTRATION NCT00164736 for BAN study.
Collapse
Affiliation(s)
- Allison S Thomas
- Department of Microbiology, Boston University School of Medicine, Boston, United States of America
| | - Carolyn Coote
- Department of Medicine, Boston Medical Center, Boston, United States of America
| | - Yvetane Moreau
- Department of Medicine, Boston Medical Center, Boston, United States of America
| | - John E Isaac
- Department of Medicine, Boston Medical Center, Boston, United States of America
| | - Alexander C Ewing
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, United States of America
| | - Athena P Kourtis
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, United States of America
| | - Manish Sagar
- Department of Medicine, Boston Medical Center, Boston, United States of America
| |
Collapse
|
9
|
Ebonwu J, Lassaunière R, Paximadis M, Goosen M, Strehlau R, Gray GE, Kuhn L, Tiemessen CT. An HIV Vaccine Protective Allele in FCGR2C Associates With Increased Odds of Perinatal HIV Acquisition. Front Immunol 2021; 12:760571. [PMID: 34917081 PMCID: PMC8668943 DOI: 10.3389/fimmu.2021.760571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
In the Thai RV144 HIV-1 vaccine trial, a three-variant haplotype within the Fc gamma receptor 2C gene (FCGR2C) reduced the risk of HIV-1 acquisition. A follow-on trial, HVTN702, of a similar vaccine candidate found no efficacy in South Africa, where the predominant population is polymorphic for only a single variant in the haplotype, c.134-96C>T (rs114945036). To investigate a role for this variant in HIV-1 acquisition in South Africans, we used the model of maternal-infant HIV-1 transmission. A nested case-control study was conducted of infants born to mothers living with HIV-1, comparing children with perinatally-acquired HIV-1 (cases, n = 176) to HIV-1-exposed uninfected children (controls, n = 349). All had received nevirapine for prevention of mother-to-child transmission. The FCGR2C copy number and expression variants (c.-386G>C, c.-120A>T c.169T>C, and c.798+1A>G) were determined using a multiplex ligation-dependent probe amplification assay and the c.134-96C>T genotype with Sanger sequencing. The copy number, genotype and allele carriage were compared between groups using univariate and multivariate logistic regression. The FCGR2C c.134-96C>T genotype distribution and copy number differed significantly between HIV-1 cases and exposed-uninfected controls (P = 0.002, P Bonf = 0.032 and P = 0.010, P Bonf = > 0.05, respectively). The FCGR2C c.134-96T allele was overrepresented in the cases compared to the controls (58% vs 42%; P = 0.001, P Bonf = 0.016). Adjusting for birthweight and FCGR2C copy number, perinatal HIV-1 acquisition was associated with the c.134-96C>T (AOR = 1.89; 95% CI 1.25-2.87; P = 0.003, P Bonf = 0.048) and c.169C>T (AOR = 2.39; 95% CI 1.45-3.95; P = 0.001, P Bonf = 0.016) minor alleles but not the promoter variant at position c.-386G>C. The c.134-96C>T variant was in strong linkage disequilibrium with the c.169C>T variant, but remained significantly associated with perinatal acquisition when adjusted for c.169C>T in multivariate analysis. In contrast to the protective effect observed in the Thai RV144 trial, we found the FCGR2C variant c.134-96T-allele associated with increased odds of perinatal HIV-1 acquisition in South African children. These findings, taken together with a similar deleterious association found with HIV-1 disease progression in South African adults, highlight the importance of elucidating the functional relevance of this variant in different populations and vaccination/disease contexts.
Collapse
Affiliation(s)
- Joy Ebonwu
- Division of Public Health Surveillance and Response, National Institute for Communicable Diseases, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ria Lassaunière
- Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | - Maria Paximadis
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Centre for HIV & STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Mark Goosen
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Centre for HIV & STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Renate Strehlau
- Empilweni Services and Research Unit, Rahima Moosa Mother and Child Hospital, Johannesburg, South Africa.,Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Glenda E Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,South African Medical Research Council, Cape Town, South Africa
| | - Louise Kuhn
- Gertrude H. Sergievsky Centre, College of Physicians and Surgeons, Columbia University, New York, NY, United States.,Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Caroline T Tiemessen
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Centre for HIV & STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
| |
Collapse
|
10
|
Sobia P, Pillay T, Liebenberg LJP, Sivro A, Mansoor LE, Osman F, Passmore JAS, Abdool Karim Q, Abdool Karim SS, Baxter C, McKinnon LR, Archary D. Higher mucosal antibody concentrations in women with genital tract inflammation. Sci Rep 2021; 11:23514. [PMID: 34873252 PMCID: PMC8648917 DOI: 10.1038/s41598-021-02954-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/18/2021] [Indexed: 11/08/2022] Open
Abstract
Inflammatory cytokines augment humoral responses by stimulating antibody production and inducing class-switching. In women, genital inflammation (GI) significantly modifies HIV risk. However, the impact of GI on mucosal antibodies remains undefined. We investigated the impact of GI, pre-HIV infection, on antibody isotypes and IgG subclasses in the female genital tract. Immunoglobulin (Ig) isotypes, IgG subclasses and 48 cytokines were measured prior to HIV infection in cervicovaginal lavages (CVL) from 66 HIV seroconverters (cases) and 66 matched HIV-uninfected women (controls) enrolled in the CAPRISA 004 and 008 1% tenofovir gel trials. Pre-HIV infection, cases had significantly higher genital IgM (4.13; IQR, 4.04-4.19) compared to controls (4.06; IQR, 3.90-4.20; p = 0.042). More than one-quarter of cases (27%) had GI compared to just over one-tenth (12%) in controls. Significantly higher IgG1, IgG3, IgG4 and IgM (all p < 0.05) were found in women stratified for GI compared to women without. Adjusted linear mixed models showed several pro-inflammatory, chemotactic, growth factors, and adaptive cytokines significantly correlated with higher titers of IgM, IgA and IgG subclasses (p < 0.05). The strong and significant positive correlations between mucosal antibodies and markers of GI suggest that GI may impact mucosal antibody profiles. These findings require further investigation to establish a plausible biological link between the local inflammatory milieu and its consequence on these genital antibodies.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
| | - Thevani Pillay
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
| | - Lenine J P Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
- Department of Medical Microbiology, University of Kwazulu-Natal, Durban, South Africa
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
- Department of Medical Microbiology, University of Kwazulu-Natal, Durban, South Africa
| | - Leila E Mansoor
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
| | - Farzana Osman
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
| | - Jo-Ann S Passmore
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
- Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Cheryl Baxter
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
| | - Lyle R McKinnon
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- National HIV and Retrovirology Labs, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, 2nd Floor, Doris Duke Medical Research Institute, 719 Umbilo Road, Durban, 4041, South Africa.
- Department of Medical Microbiology, University of Kwazulu-Natal, Durban, South Africa.
| |
Collapse
|
11
|
Thomas AS, Moreau Y, Jiang W, Isaac JE, Ewing A, White LF, Kourtis AP, Sagar M. Pre-existing infant antibody-dependent cellular cytotoxicity associates with reduced HIV-1 acquisition and lower morbidity. Cell Rep Med 2021; 2:100412. [PMID: 34755132 PMCID: PMC8561235 DOI: 10.1016/j.xcrm.2021.100412] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/18/2021] [Accepted: 09/22/2021] [Indexed: 11/24/2022]
Abstract
In humans, pre-existing anti-HIV-1 neutralizing antibodies (nAbs) have not been associated with decreased HIV-1 acquisition. Here, we evaluate antibody-dependent cellular cytotoxicity (ADCC) present in pre-transmission infant and maternal plasma and breast milk (BM) against the contemporaneous maternal HIV-1 variants. HIV-1-exposed uninfected compared with HIV-1-exposed infected infants have higher ADCC and a combination of ADCC and nAb responses against their corresponding mother's strains. ADCC does not correlate with nAbs, suggesting they are independent activities. The infected infants with high ADCC compared with low ADCC, but not those with higher ADCC plus nAbs, have lower morbidity up to 1 year after birth. A higher IgA to IgG ratio, observed in BM supernatants and in a higher proportion of the infected compared with the uninfected infants, associates with lower ADCC. Against the exposure strains, ADCC, more than nAbs, associates with both lower mother-to-child transmission and decreased post-infection infant morbidity.
Collapse
Affiliation(s)
- Allison S. Thomas
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | - Yvetane Moreau
- Department of Medicine, Boston Medical Center, Boston, MA, USA
| | - Wenqing Jiang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - John E. Isaac
- Department of Medicine, Boston Medical Center, Boston, MA, USA
| | - Alexander Ewing
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Laura F. White
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Athena P. Kourtis
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Manish Sagar
- Department of Medicine, Boston Medical Center, Boston, MA, USA
| |
Collapse
|
12
|
Lamptey H, Bonney EY, Adu B, Kyei GB. Are Fc Gamma Receptor Polymorphisms Important in HIV-1 Infection Outcomes and Latent Reservoir Size? Front Immunol 2021; 12:656894. [PMID: 34017334 PMCID: PMC8129575 DOI: 10.3389/fimmu.2021.656894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Fc gamma receptors (FcγR) are cell surface glycoproteins which trigger specific effector-cell responses when cross-linked with the Fc portions of immunoglobulin (IgG) antibodies. During HIV-1 infection, the course of disease progression, ART response, and viral reservoir size vary in different individuals. Several factors may account for these differences; however, Fc gamma receptor gene polymorphisms, which influence receptor binding to IgG antibodies, are likely to play a key role. FcγRIIa (CD32) was recently reported as a potential marker for latent HIV reservoir, however, this assertion is still inconclusive. Whether FcγR polymorphisms influence the size of the viral reservoir, remains an important question in HIV cure studies. In addition, potential cure or viral suppression methods such as broadly neutralizing antibody (bNAbs) may depend on FcγRs to control the virus. Here, we discuss the current evidence on the potential role played by FcγR polymorphisms in HIV-1 infection, treatment and vaccine trial outcomes. Importantly, we highlight contrasting findings that may be due to multiple factors and the relatively limited data from African populations. We recommend further studies especially in sub-Saharan Africa to confirm the role of FcγRIIa in the establishment of latent reservoir and to determine their influence in therapies involving bNAbs.
Collapse
Affiliation(s)
- Helena Lamptey
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Evelyn Y. Bonney
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - George B. Kyei
- Department of Virology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- Department of Medicine, Washington University School of Medicine in St Louis, St. Louis, MO, United States
- Medical and Scientific Research Centre, University of Ghana Medical Centre, University of Ghana, Accra, Ghana
| |
Collapse
|
13
|
Ding Y, Kong D, Li D, Zhang Y, Hong K, Liang H, Ma L. Characterization of antibody-dependent cellular cytotoxicity induced by the plasma from persons living with HIV-1 based on target cells with or without CD4 molecules. Microbes Infect 2021; 23:104805. [PMID: 33711449 DOI: 10.1016/j.micinf.2021.104805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/13/2021] [Accepted: 03/01/2021] [Indexed: 11/30/2022]
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is essential for reducing the reservoir of latent virus in persons living with HIV-1 (PLWH). This study evaluated the plasma's ADCC activity from treatment-naïve PLWH based on target cells with or without CD4 molecules. We found that the distribution of plasma activities to mediate ADCC is different between 8E5 cells (CD4-) and NL4-3-infected CEM.NKR.CCR5 cells (CD4+). There was no correlation between the IgG-binding ability and ADCC activity. The binding ability of the 8E5 cells (2.2%) to A32 antibody was significantly lower than that of CEM.NKR.CCR5 cells (69.3%). After incubating the 8E5 cells with CD4-mimetic compound, it did not increase the binding ability with the A32 antibody. After incubation with CD4+ T cells, the binding ability of the 8E5 cells for the A32 antibody increased significantly, which implies that the conformation of the Env protein open and expose the CD4-induced epitopes. The effect of the ADCC in plasma directly applied to 8E5 cells was positively correlated with that of the NL4-3-infected CEM.NKR.CCR5 cells. In conclusion, ADCC induction in plasma was general in the treatment-naïve PLWH. The ADCC activity levels differed when target cells with or without CD4 molecules were evaluated; When designing experiments on ADCC, full consideration should be given to this immune phenomenon.
Collapse
Affiliation(s)
- Yibo Ding
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Desheng Kong
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China; Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing 100176, China
| | - Dan Li
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yuanyuan Zhang
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Kunxue Hong
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Hua Liang
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Liying Ma
- State Key Laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| |
Collapse
|
14
|
Astorga-Gamaza A, Vitali M, Borrajo ML, Suárez-López R, Jaime C, Bastus N, Serra-Peinado C, Luque-Ballesteros L, Blanch-Lombarte O, Prado JG, Lorente J, Pumarola F, Pellicer M, Falcó V, Genescà M, Puntes V, Buzon MJ. Antibody cooperative adsorption onto AuNPs and its exploitation to force natural killer cells to kill HIV-infected T cells. NANO TODAY 2021; 36:101056. [PMID: 34394703 PMCID: PMC8360327 DOI: 10.1016/j.nantod.2020.101056] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
HIV represents a persistent infection which negatively alters the immune system. New tools to reinvigorate different immune cell populations to impact HIV are needed. Herein, a novel nanotool for the specific enhancement of the natural killer (NK) immune response towards HIV-infected T-cells has been developed. Bispecific Au nanoparticles (BiAb-AuNPs), dually conjugated with IgG anti-HIVgp120 and IgG anti-human CD16 antibodies, were generated by a new controlled, linker-free and cooperative conjugation method promoting the ordered distribution and segregation of antibodies in domains. The cooperatively-adsorbed antibodies fully retained the capabilities to recognize their cognate antigen and were able to significantly enhance cell-to-cell contact between HIV-expressing cells and NK cells. As a consequence, the BiAb-AuNPs triggered a potent cytotoxic response against HIV-infected cells in blood and human tonsil explants. Remarkably, the BiAb-AuNPs were able to significantly reduce latent HIV infection after viral reactivation in a primary cell model of HIV latency. This novel molecularly-targeted strategy using a bispecific nanotool to enhance the immune system represents a new approximation with potential applications beyond HIV.
Collapse
Affiliation(s)
- Antonio Astorga-Gamaza
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Michele Vitali
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireya L. Borrajo
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rosa Suárez-López
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Carlos Jaime
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Neus Bastus
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Carla Serra-Peinado
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Luque-Ballesteros
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oscar Blanch-Lombarte
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain
| | - Julia G. Prado
- IrsiCaixa AIDS Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona, Badalona, Spain
| | - Juan Lorente
- Otorhinolaryngology Department, Vall d’Hebron University Hospital, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Felix Pumarola
- Otorhinolaryngology Department, Vall d’Hebron University Hospital, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Marc Pellicer
- Otorhinolaryngology Department, Vall d’Hebron University Hospital, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Vicenç Falcó
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Meritxell Genescà
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Víctor Puntes
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Corresponding author at: Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Maria J. Buzon
- Infectious Disease Department, Hospital Universitario Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Corresponding author. (V. Puntes), (M.J. Buzon)
| |
Collapse
|
15
|
Mouquet H. MARVellous Sidekick Antibodies. Cell Host Microbe 2020; 27:856-858. [PMID: 32526181 DOI: 10.1016/j.chom.2020.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Human monoclonal antibodies are immune weapons that hold great promise for treating Marburg virus (MARV) infection. In this issue, Ilinykh et al. unveil non-neutralizing inhibitory properties of antibodies to the Wing region of the viral spike, which alongside with neutralizers are pivotal to in vivo protection against MARV infection.
Collapse
Affiliation(s)
- Hugo Mouquet
- Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris, France; INSERM U1222, Paris, France.
| |
Collapse
|
16
|
Duchemin M, Tudor D, Cottignies-Calamarte A, Bomsel M. Antibody-Dependent Cellular Phagocytosis of HIV-1-Infected Cells Is Efficiently Triggered by IgA Targeting HIV-1 Envelope Subunit gp41. Front Immunol 2020; 11:1141. [PMID: 32582208 PMCID: PMC7296124 DOI: 10.3389/fimmu.2020.01141] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/11/2020] [Indexed: 12/25/2022] Open
Abstract
Antibodies mediate a broad array of non-neutralizing Fc-mediated functions against HIV-1 including antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Accordingly, ADCC and ADCP induced by anti-HIV envelope gp120 IgG have been correlated to the limited success of the HIV-1 phase III vaccine trial RV144. It remains elusive whether ADCP can also be triggered by IgA, the isotype predominant at mucosal surfaces through which HIV-1 is mainly transmitted. Yet, we have previously shown that the HIV envelope subunit gp41-specific broadly neutralizing antibody 2F5 under the IgA isotype (2F5-IgA) triggers ADCC and cooperates with 2F5-IgG to increase HIV-1-infected cell lysis. Here, we now demonstrate that 2F5-IgA, more efficiently than 2F5-IgG, induces ADCP not only of gp41-coated beads but also of primary HIV-1-infected cells in a FcαRI-dependent manner. Both primary monocytes and neutrophils can act as effector cells of 2F5-IgA-mediated ADCP, although with different kinetics with faster neutrophil phagocytosis. However, unlike for ADCC, 2F5-IgA and 2F5-IgG do not cooperate to increase ADCP. Altogether, our results reveal that gp41-specific IgA mediate the efficient phagocytosis of HIV-1-infected cells. Inducing such ADCC and ADCP-prone IgA response by vaccination in addition to anti-HIV envelope IgG, might increase the protection against HIV acquisition at mucosal level.
Collapse
Affiliation(s)
- Maxence Duchemin
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, CNRS UMR 8104, Paris, France.,INSERM U1016, Paris, France.,Université Paris, Paris, France
| | - Daniela Tudor
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, CNRS UMR 8104, Paris, France.,INSERM U1016, Paris, France.,Université Paris, Paris, France
| | - Andréa Cottignies-Calamarte
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, CNRS UMR 8104, Paris, France.,INSERM U1016, Paris, France.,Université Paris, Paris, France
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Cochin Institute, CNRS UMR 8104, Paris, France.,INSERM U1016, Paris, France.,Université Paris, Paris, France
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW In the absence of a protective vaccine against HIV-1, passive immunization using novel broadly neutralizing antibodies (bNAbs) is an attractive concept for HIV-1 prevention. Here, we summarize the results of preclinical and clinical studies of bNAbs, discuss strategies for optimizing bNAb efficacy and lay out current pathways for the development of bNAbs as prophylaxis. RECENT FINDINGS Passive transfer of second-generation bNAbs results inpotent protection against infection in preclinical animal models. Furthermore, multiple bNAbs targeting different epitopes on the HIV-1 envelope trimer are in clinical evaluation and have demonstrated favorable safety profiles and robust antiviral activity in chronically infected individuals. The confirmation that passive immunization with bNAb(s) will prevent HIV-1 acquisition in humans is pending and the focus of ongoing investigations. Given the global diversity of HIV-1, bNAb combinations or multispecific antibodies will most likely be required to produce the necessary breadth for effective protection. SUMMARY Encouraging results from preclinical and clinical studies support the development of bNAbs for prevention and a number of antibodies with exceptional breadth and potency are available for clinical evaluation. Further optimization of viral coverage and antibody half-life will accelerate the clinical implementation of bNAbs as a critical tool for HIV-1 prevention strategies.
Collapse
|
18
|
Thomas AS, Ghulam-Smith M, Olson A, Coote C, Gonzales O, Sagar M. A new cell line for assessing HIV-1 antibody dependent cellular cytotoxicity against a broad range of variants. J Immunol Methods 2020; 480:112766. [PMID: 32135162 DOI: 10.1016/j.jim.2020.112766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/09/2019] [Accepted: 02/25/2020] [Indexed: 01/26/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) studies suggest that antibody-dependent cellular cytotoxicity (ADCC) influences both virus acquisition and subsequent disease outcome. Technical issues with currently available assays, however, have limited the ability to comprehensively assess the impact of ADCC on transmission and disease progression. Commonly used ADCC assays use a target cell line, CEM.NKr-CCR5-Luc, that often does not support replication of relevant HIV-1 variants. Thus, the extent of ADCC responses against a large panel of HIV-1 strains often cannot be assessed using the currently available methods. We developed two new reporter cell-lines (MT4-CCR5-Luc and PM1-CCR5-Luc) to overcome these issues. MT4-CCR5-Luc cells are resistant, whereas PM1-CCR5-Luc cells are susceptible, to killing by a natural killer cell line, CD16+KHYG-1, in the absence of antibody. Polyclonal HIVIG gave similar ADCC estimates against HIV-1 isolate, NL4-3, regardless of which of the three cell lines were used as the targets. In contrast to CEM.NKr-CCR5-Luc and PM1-CCR5-Luc, however, MT4-CCR5-Luc target cells produce significantly higher luciferase after exposure to various HIV-1 strains, including transmitted founder variants and viruses incorporating specific envelopes of interest. This higher luciferase expression does not yield spurious results because ADCC estimates are similar when killing is assessed by both reporter protein expression and flow cytometry. Furthermore, ADCC estimates derived from MT4-CCR5-Luc cells are not skewed by non-antibody contents present in human plasma. In aggregate, the MT4-CCR5-Luc cell line can be used to estimate monoclonal antibody or plasma-induced ADCC responses against a diverse range of HIV-1 envelopes relevant for transmission and disease progression studies.
Collapse
Affiliation(s)
- Allison S Thomas
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
| | | | - Alex Olson
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Carolyn Coote
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Oscar Gonzales
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Manish Sagar
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
19
|
Li SS, Gilbert PB, Carpp LN, Pyo CW, Janes H, Fong Y, Shen X, Neidich SD, Goodman D, deCamp A, Cohen KW, Ferrari G, Hammer SM, Sobieszczyk ME, Mulligan MJ, Buchbinder SP, Keefer MC, DeJesus E, Novak RM, Frank I, McElrath MJ, Tomaras GD, Geraghty DE, Peng X. Fc Gamma Receptor Polymorphisms Modulated the Vaccine Effect on HIV-1 Risk in the HVTN 505 HIV Vaccine Trial. J Virol 2019; 93:e02041-18. [PMID: 31434737 PMCID: PMC6803257 DOI: 10.1128/jvi.02041-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 08/14/2019] [Indexed: 12/19/2022] Open
Abstract
HIV Vaccine Trials Network (HVTN) 505 was a phase 2b efficacy trial of a DNA/recombinant adenovirus 5 (rAd5) HIV vaccine regimen. Although the trial was stopped early for lack of overall efficacy, later correlates of risk and sieve analyses generated the hypothesis that the DNA/rAd5 vaccine regimen protected some vaccinees from HIV infection yet enhanced HIV infection risk for others. Here, we assessed whether and how host Fc gamma receptor (FcγR) genetic variations influenced the DNA/rAd5 vaccine regimen's effect on HIV infection risk. We found that vaccine receipt significantly increased HIV acquisition compared with placebo receipt among participants carrying the FCGR2C-TATA haplotype (comprising minor alleles of four FCGR2C single-nucleotide polymorphism [SNP] sites) (hazard ratio [HR] = 9.79, P = 0.035) but not among participants without the haplotype (HR = 0.86, P = 0.67); the interaction of vaccine and haplotype effect was significant (P = 0.034). Similarly, vaccine receipt increased HIV acquisition compared with placebo receipt among participants carrying the FCGR3B-AGA haplotype (comprising minor alleles of the 3 FCGR3B SNPs) (HR = 2.78, P = 0.058) but not among participants without the haplotype (HR = 0.73, P = 0.44); again, the interaction of vaccine and haplotype was significant (P = 0.047). The FCGR3B-AGA haplotype also influenced whether a combined Env-specific CD8+ T-cell polyfunctionality score and IgG response correlated significantly with HIV risk; an FCGR2A SNP and two FCGR2B SNPs influenced whether anti-gp140 antibody-dependent cellular phagocytosis correlated significantly with HIV risk. These results provide further evidence that Fc gamma receptor genetic variations may modulate HIV vaccine effects and immune function after HIV vaccination.IMPORTANCE By analyzing data from the HVTN 505 efficacy trial of a DNA/recombinant adenovirus 5 (rAd5) vaccine regimen, we found that host genetics, specifically Fc gamma receptor genetic variations, influenced whether receiving the DNA/rAd5 regimen was beneficial, neutral, or detrimental to an individual with respect to HIV-1 acquisition risk. Moreover, Fc gamma receptor genetic variations influenced immune responses to the DNA/rAd5 vaccine regimen. Thus, Fc gamma receptor genetic variations should be considered in the analysis of future HIV vaccine trials and the development of HIV vaccines.
Collapse
Affiliation(s)
- Shuying S Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Chul-Woo Pyo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Scott D Neidich
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Derrick Goodman
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Allan deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kristen W Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Scott M Hammer
- Division of Infectious Diseases, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Magdalena E Sobieszczyk
- Division of Infectious Diseases, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Mark J Mulligan
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Susan P Buchbinder
- Department of Medicine, University of California, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Michael C Keefer
- Division of Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
- Department of Immunology, Duke University, Durham, North Carolina, USA
| | - Daniel E Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
20
|
The potential of engineered antibodies for HIV-1 therapy and cure. Curr Opin Virol 2019; 38:70-80. [PMID: 31421319 DOI: 10.1016/j.coviro.2019.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 12/21/2022]
Abstract
Broadly neutralizing antibodies (bnAbs) are currently under investigation as a therapy for HIV-1 infection and recent clinical trials have shown prolonged viral suppression by bnAbs during antiretroviral treatment interruption. Interestingly, these bnAbs also showed the ability to activate the host immune system to clear HIV-1 infected cells. There are many possibilities to further increase the potential efficacy of bnAbs. Most notably, Fc domain engineering to improve half-life and increase engagement of effector cells will augment two advantages of bnAbs. Moreover, antibody engineering can improve affinity and recognition of conserved epitopes and allows the combination of multiple epitope specificities in a single molecule. These increasingly potent and broad antibodies may prove valuable as alternative HIV-1 therapeutic and possibly in curative approaches.
Collapse
|
21
|
Tolbert WD, Sherburn RT, Van V, Pazgier M. Structural Basis for Epitopes in the gp120 Cluster A Region that Invokes Potent Effector Cell Activity. Viruses 2019; 11:v11010069. [PMID: 30654465 PMCID: PMC6357199 DOI: 10.3390/v11010069] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/28/2022] Open
Abstract
While a number of therapeutic options to control the progression of human immunodeficiency virus (HIV-1) now exist, a broadly effective preventive vaccine is still not available. Through detailed structural analysis of antibodies able to induce potent effector cell activity, a number of Env epitopes have been identified which have the potential to be considered vaccine candidates. These antibodies mainly target the gp120 Cluster A region which is only exposed upon viral binding to the target cell with epitopes becoming available for antibody binding during viral entry and fusion and, therefore, after the effective window for neutralizing antibody activity. This review will discuss recent advances in the structural characterization of these important targets with a special focus on epitopes that are involved in Fc-mediated effector function without direct viral neutralizing activities.
Collapse
Affiliation(s)
- William D Tolbert
- Infectious Diseases Division, Department of Medicine of Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Rebekah T Sherburn
- Infectious Diseases Division, Department of Medicine of Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Verna Van
- Department of Biochemistry and Molecular Biology of University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Marzena Pazgier
- Infectious Diseases Division, Department of Medicine of Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| |
Collapse
|
22
|
Single-Domain Antibodies and Their Formatting to Combat Viral Infections. Antibodies (Basel) 2018; 8:antib8010001. [PMID: 31544807 PMCID: PMC6640686 DOI: 10.3390/antib8010001] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Since their discovery in the 1990s, single-domain antibodies (VHHs), also known as Nanobodies®, have changed the landscape of affinity reagents. The outstanding solubility, stability, and specificity of VHHs, as well as their small size, ease of production and formatting flexibility favor VHHs over conventional antibody formats for many applications. The exceptional ease by which it is possible to fuse VHHs with different molecular modules has been particularly explored in the context of viral infections. In this review, we focus on VHH formats that have been developed to combat viruses including influenza viruses, human immunodeficiency virus-1 (HIV-1), and human respiratory syncytial virus (RSV). Such formats may significantly increase the affinity, half-life, breadth of protection of an antiviral VHH and reduce the risk of viral escape. In addition, VHHs can be equipped with effector functions, for example to guide components of the immune system with high precision to sites of viral infection.
Collapse
|
23
|
Ren Y, Korom M, Truong R, Chan D, Huang SH, Kovacs CC, Benko E, Safrit JT, Lee J, Garbán H, Apps R, Goldstein H, Lynch RM, Jones RB. Susceptibility to Neutralization by Broadly Neutralizing Antibodies Generally Correlates with Infected Cell Binding for a Panel of Clade B HIV Reactivated from Latent Reservoirs. J Virol 2018; 92:e00895-18. [PMID: 30209173 PMCID: PMC6232479 DOI: 10.1128/jvi.00895-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
Efforts to cure human immunodeficiency virus (HIV) infection are obstructed by reservoirs of latently infected CD4+ T cells that can reestablish viremia. HIV-specific broadly neutralizing antibodies (bNAbs), defined by unusually wide neutralization breadths against globally diverse viruses, may contribute to the elimination of these reservoirs by binding to reactivated cells, thus targeting them for immune clearance. However, the relationship between neutralization of reservoir isolates and binding to corresponding infected primary CD4+ T cells has not been determined. Thus, the extent to which neutralization breadths and potencies can be used to infer the corresponding parameters of infected cell binding is currently unknown. We assessed the breadths and potencies of bNAbs against 36 viruses reactivated from peripheral blood CD4+ T cells from antiretroviral (ARV)-treated HIV-infected individuals by using paired neutralization and infected cell binding assays. Single-antibody breadths ranged from 0 to 64% for neutralization (80% inhibitory concentration [IC80] of ≤10 μg/ml) and from 0 to 89% for binding, with two-antibody combinations (results for antibody combinations are theoretical/predicted) reaching levels of 0 to 83% and 50 to 100%, respectively. Infected cell binding correlated with virus neutralization for 10 of 14 antibodies (e.g., for 3BNC117, r = 0.82 and P < 0.0001). Heterogeneity was observed, however, with a lack of significant correlation for 2G12, CAP256.VRC26.25, 2F5, and 4E10. Our results provide guidance on the selection of bNAbs for interventional cure studies, both by providing a direct assessment of intra- and interindividual variabilities in neutralization and infected cell binding in a novel cohort and by defining the relationships between these parameters for a panel of bNAbs.IMPORTANCE Although antiretroviral therapies have improved the lives of people who are living with HIV, they do not cure infection. Efforts are being directed towards harnessing the immune system to eliminate the virus that persists, potentially resulting in virus-free remission without medication. HIV-specific antibodies hold promise for such therapies owing to their ability to both prevent the infection of new cells (neutralization) and direct the killing of infected cells. We isolated 36 HIV strains from individuals whose virus was suppressed by medication and tested 14 different antibodies for neutralization of these viruses and for binding to cells infected with the same viruses (critical for engaging natural killer cells). For both neutralization and infected cell binding, we observed variation both between individuals and amongst different viruses within an individual. For most antibodies, neutralization activity correlated with infected cell binding. These data provide guidance on the selection of antibodies for clinical trials.
Collapse
Affiliation(s)
- Yanqin Ren
- Division of Infectious Diseases, Weill Cornell Medical College, New York, New York, USA
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Maria Korom
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Ronald Truong
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Dora Chan
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Szu-Han Huang
- Division of Infectious Diseases, Weill Cornell Medical College, New York, New York, USA
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | | | - Erika Benko
- Maple Leaf Medical Clinic, Toronto, Ontario, Canada
| | | | - John Lee
- NantBioScience Inc./NantKwest LLC, Culver City, California, USA
| | | | - Richard Apps
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Harris Goldstein
- Department of Pediatrics, Albert Einstein College of Medicine, New York, New York, USA
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Rebecca M Lynch
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - R Brad Jones
- Division of Infectious Diseases, Weill Cornell Medical College, New York, New York, USA
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
24
|
Thida W, Kuwata T, Maeda Y, Yamashiro T, Tran GV, Nguyen KV, Takiguchi M, Gatanaga H, Tanaka K, Matsushita S. The role of conventional antibodies targeting the CD4 binding site and CD4-induced epitopes in the control of HIV-1 CRF01_AE viruses. Biochem Biophys Res Commun 2018; 508:46-51. [PMID: 30470571 DOI: 10.1016/j.bbrc.2018.11.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 11/12/2018] [Indexed: 12/21/2022]
Abstract
HIV-1 CRF01_AE viruses are highly prevalent in Southeast Asia. However, vulnerability sites in Env of CRF01_AE viruses have not been investigated sufficiently. We examined the sensitivity of CRF01_AE viruses from Japan and Vietnam, together with subtype B viruses from Japan, to neutralization and Fc-mediated signaling. Neutralization coverage of broadly neutralizing antibodies (bnAbs), 2G12 and b12, was significantly low against CRF01_AE viruses, compared with subtype B viruses. In contrast, the conventional antibody targeting the CD4 binding site (CD4bs), 49G2, showed better neutralization and Fc-mediated signaling activities against CRF01_AE viruses than subtype B viruses. Fc-mediated signaling activity of anti-CD4 induced (CD4i) antibody, 4E9C, was also detected against CRF01_AE viruses more than subtype B viruses. These results suggest that conventional antibodies against CD4bs and CD4i may play an important role in the control of CRF01_AE viruses.
Collapse
Affiliation(s)
- Win Thida
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Takeo Kuwata
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan.
| | - Yosuke Maeda
- Department of Microbiology, Kumamoto University, Kumamoto, Japan
| | - Tetsu Yamashiro
- Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Giang Van Tran
- National Hospital of Tropical Diseases, Dong Da District, Hanoi, Viet Nam
| | - Kinh Van Nguyen
- National Hospital of Tropical Diseases, Dong Da District, Hanoi, Viet Nam
| | | | - Hiroyuki Gatanaga
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kazuki Tanaka
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
25
|
Richardson SI, Chung AW, Natarajan H, Mabvakure B, Mkhize NN, Garrett N, Abdool Karim S, Moore PL, Ackerman ME, Alter G, Morris L. HIV-specific Fc effector function early in infection predicts the development of broadly neutralizing antibodies. PLoS Pathog 2018; 14:e1006987. [PMID: 29630668 PMCID: PMC5908199 DOI: 10.1371/journal.ppat.1006987] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/19/2018] [Accepted: 03/22/2018] [Indexed: 12/24/2022] Open
Abstract
While the induction of broadly neutralizing antibodies (bNAbs) is a major goal of HIV vaccination strategies, there is mounting evidence to suggest that antibodies with Fc effector function also contribute to protection against HIV infection. Here we investigated Fc effector functionality of HIV-specific IgG plasma antibodies over 3 years of infection in 23 individuals, 13 of whom developed bNAbs. Antibody-dependent cellular phagocytosis (ADCP), complement deposition (ADCD), cellular cytotoxicity (ADCC) and cellular trogocytosis (ADCT) were detected in almost all individuals with levels of activity increasing over time. At 6 months post-infection, individuals with bNAbs had significantly higher levels of ADCD and ADCT that correlated with antibody binding to C1q and FcγRIIa respectively. In addition, antibodies from individuals with bNAbs showed more IgG subclass diversity to multiple HIV antigens which also correlated with Fc polyfunctionality. Germinal center activity represented by CXCL13 levels and expression of activation-induced cytidine deaminase (AID) was found to be associated with neutralization breadth, Fc polyfunctionality and IgG subclass diversity. Overall, multivariate analysis by random forest classification was able to group bNAb individuals with 85% sensitivity and 80% specificity based on the properties of their antibody Fc early in HIV infection. Thus, the Fc effector function profile predicted the development of neutralization breadth in this cohort, suggesting that intrinsic immune factors within the germinal center provide a mechanistic link between the Fc and Fab of HIV-specific antibodies. Some HIV-infected individuals develop antibodies that are capable of neutralizing the majority of HIV strains, a highly desirable function mediated by the antibody Fab portion. While antibodies elicited by current vaccines have failed to recreate this activity, the partial protection seen in the RV144 vaccine trial has been attributed to antibody Fc-mediated effector functions such as cell killing. In this study, we found that HIV-infected individuals who show a diversified and potent Fc response early in infection were more likely to develop broadly neutralizing antibodies later on. Examination of B cell functions associated with good germinal center activity, provided evidence for a common mechanistic link between the regulation of the Fc and Fab mediated activities in these individuals. Our finding of an Fc effector function profile that arises early and predicts neutralization breadth could be used in the evaluation of vaccine candidates designed to generate neutralizing antibodies. Common immune determinants associated with both Fab and Fc function could furthermore be exploited for vaccine design to harness the full potential of HIV-specific antibodies.
Collapse
Affiliation(s)
- Simone I. Richardson
- Centre for HIV and STI’s, National Institute for Communicable Diseases, Johannesburg, Gauteng, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Harini Natarajan
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Batsirai Mabvakure
- Centre for HIV and STI’s, National Institute for Communicable Diseases, Johannesburg, Gauteng, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Nonhlanhla N. Mkhize
- Centre for HIV and STI’s, National Institute for Communicable Diseases, Johannesburg, Gauteng, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu Natal, South Africa
| | - Salim Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu Natal, South Africa
| | - Penny L. Moore
- Centre for HIV and STI’s, National Institute for Communicable Diseases, Johannesburg, Gauteng, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu Natal, South Africa
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Lynn Morris
- Centre for HIV and STI’s, National Institute for Communicable Diseases, Johannesburg, Gauteng, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu Natal, South Africa
- * E-mail:
| |
Collapse
|
26
|
Wines BD, Billings H, Mclean MR, Kent SJ, Hogarth PM. Antibody Functional Assays as Measures of Fc Receptor-Mediated Immunity to HIV - New Technologies and their Impact on the HIV Vaccine Field. Curr HIV Res 2018; 15:202-215. [PMID: 28322167 PMCID: PMC5543561 DOI: 10.2174/1570162x15666170320112247] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 12/23/2022]
Abstract
Background: There is now intense interest in the role of HIV-specific antibodies and the engagement of FcγR functions in the control and prevention of HIV infection. The analyses of the RV144 vaccine trial, natural progression cohorts, and macaque models all point to a role for Fc-dependent effector functions, such as cytotoxicity (ADCC) or phagocytosis (ADCP), in the control of HIV. However, reliable assays that can be reproducibly used across different laboratories to measure Fc-dependent functions, such as antibody dependent cellular cytotoxicity (ADCC) are limited. Method: This brief review highlights the importance of Fc properties for immunity to HIV, particular-ly via FcγR diversity and function. We discuss assays used to study FcR mediated functions of HIV-specific Ab, including our recently developed novel cell-free ELISA using homo-dimeric FcγR ecto-domains to detect functionally relevant viral antigen-specific antibodies. Results: The binding of these dimeric FcγR ectodomains, to closely spaced pairs of IgG Fc, mimics the engagement and cross-linking of Fc receptors by IgG opsonized virions or infected cells as the es-sential prerequisite to the induction of Ab-dependent effector functions. The dimeric FcγR ELISA reli-ably correlates with ADCC in patient responses to influenza. The assay is amenable to high throughput and could be standardized across laboratories. Conclusion: We propose the assay has broader implications for the evaluation of the quality of anti-body responses in viral infections and for the rapid evaluation of responses in vaccine development campaigns for HIV and other viral infections.
Collapse
Affiliation(s)
- Bruce D Wines
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| | - Hugh Billings
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia
| | - Milla R Mclean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Parkville, Victoria, Australia.,Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Victoria, Australia
| | - P Mark Hogarth
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
27
|
Davis-Gardner ME, Gardner MR, Alfant B, Farzan M. eCD4-Ig promotes ADCC activity of sera from HIV-1-infected patients. PLoS Pathog 2017; 13:e1006786. [PMID: 29253851 PMCID: PMC5749896 DOI: 10.1371/journal.ppat.1006786] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/02/2018] [Accepted: 12/03/2017] [Indexed: 12/17/2022] Open
Abstract
Antibody-dependent cell-mediated cytotoxity (ADCC) can eliminate HIV-1 infected cells, and may help reduce the reservoir of latent virus in infected patients. Sera of HIV-1 positive individuals include a number of antibodies that recognize epitopes usually occluded on HIV-1 envelope glycoprotein (Env) trimers. We have recently described eCD4-Ig, a potent and exceptionally broad inhibitor of HIV-1 entry that can be used to protect rhesus macaques from multiple high-dose challenges with simian-human immunodeficiency virus AD8 (SHIV-AD8). Here we show that eCD4-Ig bearing an IgG1 Fc domain (eCD4-IgG1) can mediate efficient ADCC activity against HIV-1 isolates with differing tropisms, and that it does so at least 10-fold more efficiently than CD4-Ig, even when more CD4-Ig molecules bound cell surface-expressed Env. An ADCC-inactive IgG2 form of eCD4-Ig (eCD4-IgG2) exposes V3-loop and CD4-induced epitopes on cell-expressed trimers, and renders HIV-1-infected cells susceptible to ADCC mediated by antibodies of these classes. Moreover, eCD4-IgG2, but not IgG2 forms of the broadly neutralizing antibodies VRC01 and 10-1074, enhances the ADCC activities of serum antibodies from patients by 100-fold, and significantly enhanced killing of two latently infected T-cell lines reactivated by vorinostat or TNFα. Thus eCD4-Ig is qualitatively different from CD4-Ig or neutralizing antibodies in its ability to mediate ADCC, and it may be uniquely useful in treating HIV-1 infection or reducing the reservoir of latently infected cells.
Collapse
Affiliation(s)
- Meredith E. Davis-Gardner
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Matthew R. Gardner
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Barnett Alfant
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Michael Farzan
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, United States of America
| |
Collapse
|
28
|
Mayr LM, Su B, Moog C. Non-Neutralizing Antibodies Directed against HIV and Their Functions. Front Immunol 2017; 8:1590. [PMID: 29209323 PMCID: PMC5701973 DOI: 10.3389/fimmu.2017.01590] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/06/2017] [Indexed: 12/21/2022] Open
Abstract
B cells produce a plethora of anti-HIV antibodies (Abs) but only few of them exhibit neutralizing activity. This was long considered a profound limitation for the enforcement of humoral immune responses against HIV-1 infection, especially since these neutralizing Abs (nAbs) are extremely difficult to induce. However, increasing evidence shows that additional non-neutralizing Abs play a significant role in decreasing the viral load, leading to partial and sometimes even total protection. Mechanisms suspected to participate in protection are numerous. They involve the Fc domain of Abs as well as their Fab part, and consequently the induced Ab isotype will be determinant for their functions, as well as the quantity and quality of the Fc-receptors (FcRs) expressed on immune cells. Fc-mediated inhibitory functions, such as Ab-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, aggregation, and even immune activation have been proposed. However, as for nAbs, the non-neutralizing activities are limited to a subset of anti-HIV Abs. An improved in-depth characterization of the Abs displaying these functional responses is required for the development of new vaccination strategies, which aim to selectively trigger the B cells able to induce the right functional Ab combinations both at the right place and at the right time. This review summarizes our current knowledge on non-neutralizing functional inhibitory Abs and discusses the potential benefit of inducing them via vaccination. We also provide new insight into the roles of the FcγR-mediated Ab therapeutics in clinical trials for HIV diseases.
Collapse
Affiliation(s)
- Luzia M Mayr
- INSERM U1109, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing You'an Hospital, Capital Medical University, Beijing, China
| | - Christiane Moog
- INSERM U1109, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| |
Collapse
|
29
|
Abstract
Despite major advances in antiretroviral therapy against HIV-1, an effective HIV vaccine is urgently required to reduce the number of new cases of HIV infections in the world. Vaccines are the ultimate tool in the medical arsenal to control and prevent the spread of infectious diseases such as HIV/AIDS. Several failed phase-IIb to –III clinical vaccine trials against HIV-1 in the past generated a plethora of information that could be used for better designing of an effective HIV vaccine in the future. Most of the tested vaccine candidates produced strong humoral responses against the HIV proteins; however, failed to protect due to: 1) the low levels and the narrow breadth of the HIV-1 neutralizing antibodies and the HIV-specific antibody-dependent Fc-mediated effector activities, 2) the low levels and the poor quality of the anti-HIV T-cell responses, and 3) the excessive responses to immunodominant non-protective HIV epitopes, which in some cases blocked the protective immunity and/or enhanced HIV infection. The B-cell epitopes on HIV for producing broadly neutralizing antibodies (bNAbs) against HIV have been extensively characterized, and the next step is to develop bNAb epitope immunogen for HIV vaccine. The bNAb epitopes are often conformational epitopes and therefore more difficult to construct as vaccine immunogen and likely to include immunodominant non-protective HIV epitopes. In comparison, T-cell epitopes are short linear peptides which are easier to construct into vaccine immunogen free of immunodominant non-protective epitopes. However, its difficulty lies in identifying the T-cell epitopes conserved among HIV subtypes and induce long-lasting, potent polyfunctional T-cell and cytotoxic T lymphocyte (CTL) activities against HIV. In addition, these protective T-cell epitopes must be recognized by the HLA prevalent in the country(s) targeted for the vaccine trial. In conclusion, extending from the findings from previous vaccine trials, future vaccines should combine both T- and B-cell epitopes as vaccine immunogen to induce multitude of broad and potent immune effector activities required for sterilizing protection against global HIV subtypes.
Collapse
Affiliation(s)
- Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - Cuong Q Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| | - Janet K Yamamoto
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611-0880, USA
| |
Collapse
|
30
|
Abstract
The scale and scope of the global epidemic, coupled to challenges with traditional vaccine development approaches, point toward a need for novel methodologies for HIV vaccine research. While the development of vaccines able to induce broadly neutralizing antibodies remains the ultimate goal, to date, vaccines continue to fail to induce these rare humoral immune responses. Conversely, growing evidence across vaccine platforms in both non-human primates and humans points to a role for polyclonal vaccine-induced antibody responses in protection from infection. These candidate vaccines, despite employing disparate viral vectors and immunization strategies, consistently identify a role for functional or non-traditional antibody activities as correlates of immunity. However, the precise mechanism(s) of action of these "binding" antibodies, their specific characteristics, and their ability to be selectively induced and/or potentiated to result in complete protection merits parallel investigation to neutralizing antibody-based vaccine design approaches. Ultimately, while neutralizing and functional antibody-based vaccine strategies need not be mutually exclusive, defining the specific characteristics of "protective" functional antibodies may provide a target immune profile to potentially induce more robust immunity against HIV. Specifically, one approach to guide the development of functional antibody-based vaccine strategies, termed "systems serology", offers an unbiased and comprehensive approach to systematically survey humoral immune responses, capturing the array of functions and humoral response characteristics that may be induced following vaccination with high resolution. Coupled to machine learning tools, large datasets that explore the "antibody-ome" offer a means to step back from anticipated correlates and mechanisms of protection and toward a more fundamental understanding of coordinated aspects of humoral immune responses, to more globally differentiate among vaccine candidates, and most critically, to identify the features of humoral immunity that distinguish protective from non-protective responses. Overall, the systematic serological approach described here aimed at broadly capturing the enormous biodiversity in antibody profiles that may emerge following vaccination, complements the existing cutting edge tools in the cellular immunology space that survey vaccine-induced polyfunctional cellular activity by flow cytometry, transcriptional profiling, epigenetic, and metabolomic analysis to offer a means to develop both a more nuanced and a more complete understanding of correlates of protection to support the design of functional vaccine strategies.
Collapse
Affiliation(s)
| | - Dan H Barouch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Beth Israel Deaconness Medical Center, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
31
|
Yaseen MM, Yaseen MM, Alqudah MA. Broadly neutralizing antibodies: An approach to control HIV-1 infection. Int Rev Immunol 2016; 36:31-40. [PMID: 27739924 DOI: 10.1080/08830185.2016.1225301] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although available antiretroviral therapy (ART) has changed human immunodeficiency virus (HIV)-1 infection to a non-fatal chronic disease, the economic burden of lifelong therapy, severe adverse ART effects, daily ART adherence, and emergence of ART-resistant HIV-1 mutants require prospecting for alternative therapeutic modalities. Indeed, a growing body of evidence suggests that broadly neutralizing anti-HIV-1 antibodies (BNAbs) may offer one such feasible alternative. To evaluate their therapeutic potential in established HIV-1 infection, we sought to address recent advances in pre-clinical and clinical investigations in this area of HIV-1 research. In addition, we addressed the obstacles that may impede the success of such immunotherapeutic approach, suggested strategic solutions, and briefly compared this approach with the currently used ART to open new insights for potential future passive immunotherapy for HIV-1 infection.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- a Department of Medical Laboratory Sciences , College of Applied Medical Sciences, Jordan University of Science and Technology , Irbid , Jordan
| | - Mohammad Mahmoud Yaseen
- b Department of Public Health, College of Nursing , University of Benghazi , Benghazi , Libya
| | - Mohammad Ali Alqudah
- c Department of Clinical Pharmacy , College of Pharmacy, Jordan University of Science and Technology , Irbid , Jordan
| |
Collapse
|
32
|
Affiliation(s)
- P Mark Hogarth
- Centre for Biomedicine, Burnet Institute, Melbourne, Vic., Australia.,Department of Immunology, Monash University, Melbourne, Vic., Australia.,Department of Pathology, University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
33
|
Hua CK, Ackerman ME. Engineering broadly neutralizing antibodies for HIV prevention and therapy. Adv Drug Deliv Rev 2016; 103:157-173. [PMID: 26827912 DOI: 10.1016/j.addr.2016.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 01/15/2023]
Abstract
A combination of advances spanning from isolation to delivery of potent HIV-specific antibodies has begun to revolutionize understandings of antibody-mediated antiviral activity. As a result, the set of broadly neutralizing and highly protective antibodies has grown in number, diversity, potency, and breadth of viral recognition and neutralization. These antibodies are now being further enhanced by rational engineering of their anti-HIV activities and coupled to cutting edge gene delivery and strategies to optimize their pharmacokinetics and biodistribution. As a result, the prospects for clinical use of HIV-specific antibodies to treat, clear, and prevent HIV infection are gaining momentum. Here we discuss the diverse methods whereby antibodies are being optimized for neutralization potency and breadth, biodistribution, pharmacokinetics, and effector function with the aim of revolutionizing HIV treatment and prevention options.
Collapse
|
34
|
Lu CL, Murakowski DK, Bournazos S, Schoofs T, Sarkar D, Halper-Stromberg A, Horwitz JA, Nogueira L, Golijanin J, Gazumyan A, Ravetch JV, Caskey M, Chakraborty AK, Nussenzweig MC. Enhanced clearance of HIV-1-infected cells by broadly neutralizing antibodies against HIV-1 in vivo. Science 2016; 352:1001-4. [PMID: 27199430 DOI: 10.1126/science.aaf1279] [Citation(s) in RCA: 276] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/11/2016] [Indexed: 01/09/2023]
Abstract
Antiretroviral drugs and antibodies limit HIV-1 infection by interfering with the viral life cycle. In addition, antibodies also have the potential to guide host immune effector cells to kill HIV-1-infected cells. Examination of the kinetics of HIV-1 suppression in infected individuals by passively administered 3BNC117, a broadly neutralizing antibody, suggested that the effects of the antibody are not limited to free viral clearance and blocking new infection but also include acceleration of infected cell clearance. Consistent with these observations, we find that broadly neutralizing antibodies can target CD4(+) T cells infected with patient viruses and can decrease their in vivo half-lives by a mechanism that requires Fcγ receptor engagement in a humanized mouse model. The results indicate that passive immunotherapy can accelerate elimination of HIV-1-infected cells.
Collapse
Affiliation(s)
- Ching-Lan Lu
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA. Weill Cornell Medical College, New York, NY 10065, USA
| | - Dariusz K Murakowski
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Till Schoofs
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Debolina Sarkar
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Joshua A Horwitz
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Lilian Nogueira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jovana Golijanin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Arup K Chakraborty
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA. Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA. Howard Hughes Medical Institute.
| |
Collapse
|
35
|
Abstract
As the 2014-15 Ebola virus epidemic in West Africa evolved from emergency to lesson, developers of both vaccines and therapeutic antibodies were left with the puzzlement of what kinds of anti-Ebola antibodies are predictably desirable in treating the afflicted, and what antibodies might account for the specific and lasting protection elicited by the more effective vaccines. The facile answer in virology is that neutralizing antibody (NAb) is desired and required. However, with Ebola and other filoviruses (as with many prior viral examples), there are multiple discordances in which neutralizing antibodies fail to protect animals, and others in which antibody-mediated protection is observed in the absence of measured virus neutralization. Explanation presumably resides in the protective role of antibodies that bind and functionally 'target' virus-infected cells, here called 'cell-targeting antibody', or CTAb. To be clear, many NAbs are also CTAbs, and in the case of Ebola the great majority of NAbs are likely CTAbs. Isotype, glycosylation, and other features of CTAbs are likely crucial in their capacity to mediate protection. Overall, results and analysis invite an increasingly complex view of antibody-mediated immunity to enveloped viruses.
Collapse
Affiliation(s)
- Alan Schmaljohn
- Microbiology & Immunology, University of Maryland School of Medicine,
USA,Corresponding author: Department of Microbiology & Immunology, University
of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA. Tel:
410-706-3059
| | - George K. Lewis
- Institute of Human Virology, University of Maryland School of Medicine, 725
W. Lombard St., Baltimore, Maryland, 21201, USA
| |
Collapse
|