1
|
Slein MD, Backes IM, Kelkar NS, Garland CR, Khanwalkar US, Sholukh AM, Johnston CM, Leib DA, Ackerman ME. Improving antibody-mediated protection against HSV infection by eliminating interactions with the viral Fc receptor gE/gI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624598. [PMID: 39605495 PMCID: PMC11601663 DOI: 10.1101/2024.11.20.624598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Herpes simplex virus (HSV) encodes surface glycoproteins that are host defense evasion molecules, allowing the virus to escape immune clearance. In addition to their role in neuropathogenesis and cell-cell spread, glycoproteins E and I (gE/gI) form a viral Fc receptor (vFcR) for most subclasses and allotypes of human IgG and promote evasion of humoral immune responses. While monoclonal antibodies (mAbs) protect mice from neonatal HSV (nHSV) infections, the impact of the vFcR on mAb-mediated protection by binding to IgG is unknown. Using HSV-1 with intact and ablated gE-mediated IgG Fc binding, and Fc-engineered antibodies with modified ability to interact with gE/gI, we investigated the role of the vFcR in viral pathogenesis and mAb-mediated protection from nHSV. The gD-specific human mAb HSV8 modified to lack binding to gE exhibited enhanced neutralization and in vivo protection compared to its native IgG1 form. This improved protection by the engineered mAbs was dependent on the presence of the vFcR. Human IgG3 allotypes lacking vFcR binding also exhibited enhanced antiviral activity in vivo, suggesting that vaccines that robustly induce IgG3 responses could show enhanced protection. suggesting the value of vaccination strategies that robustly induce this subclass. Lastly, analysis of longitudinal responses to acute primary genital infection in humans raised the possibility that unlike most viruses, HSV may exhibited slow induction of IgG3. In summary, this study demonstrates that mAbs lacking the ability to interact with the vFcR can exhibit improved protection from HSV-offering new prospects for antibody-based interventions.
Collapse
Affiliation(s)
- Matthew D. Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Iara M. Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | | | | - Christine M. Johnston
- Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Departments of Medicine and Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104, USA
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
2
|
Hederman AP, Remmel CA, Sharma S, Natarajan H, Weiner JA, Wrapp D, Donner C, Delforge ML, d’Angelo P, Furione M, Fornara C, McLellan JS, Lilleri D, Marchant A, Ackerman ME. Discrimination of primary and chronic cytomegalovirus infection based on humoral immune profiles in pregnancy. J Clin Invest 2024; 134:e180560. [PMID: 39207860 PMCID: PMC11473158 DOI: 10.1172/jci180560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUNDMost humans have been infected with cytomegalovirus (CMV) by midlife without clinical signs of disease. However, in settings in which the immune system is undeveloped or compromised, the virus is not adequately controlled and consequently presents a major infectious cause of both congenital disease during pregnancy as well as opportunistic infection in children and adults. With clear evidence that risk to the fetus varies with gestational age at the time of primary maternal infection, further research on humoral responses to primary CMV infection during pregnancy is needed.METHODSHere, systems serology tools were applied to characterize antibody responses to CMV infection in pregnant and nonpregnant women experiencing either primary or chronic infection.RESULTSWhereas strikingly different antibody profiles were observed depending on infection status, limited differences were associated with pregnancy status. Beyond known differences in IgM responses used clinically for identification of primary infection, distinctions observed in IgA and FcγR-binding antibodies and among antigen specificities accurately predicted infection status. Machine learning was used to define the transition from primary to chronic states and predict time since infection with high accuracy. Humoral responses diverged over time in an antigen-specific manner, with IgG3 responses toward tegument decreasing over time as typical of viral infections, while those directed to pentamer and glycoprotein B were lower during acute and greatest during chronic infection.CONCLUSIONIn sum, this work provides insights into the antibody response associated with CMV infection status in the context of pregnancy, revealing aspects of humoral immunity that have the potential to improve CMV diagnostics.FUNDINGCYMAF consortium and NIH NIAID.
Collapse
Affiliation(s)
- Andrew P. Hederman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | | | - Shilpee Sharma
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire, USA
| | - Joshua A. Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas, Austin, Texas, USA
| | - Catherine Donner
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), CUB Hôpital Erasme, Department of Obstetrics and Gynecology, Brussels, Belgium
| | - Marie-Luce Delforge
- ULB, H.U.B., CUB Hôpital Erasme, National Reference Center for Congenital Infections, Brussels, Belgium
| | - Piera d’Angelo
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Milena Furione
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Fornara
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas, Austin, Texas, USA
| | - Daniele Lilleri
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire, USA
| |
Collapse
|
3
|
Pirker AL, Vogl T. Development of systemic and mucosal immune responses against gut microbiota in early life and implications for the onset of allergies. FRONTIERS IN ALLERGY 2024; 5:1439303. [PMID: 39086886 PMCID: PMC11288972 DOI: 10.3389/falgy.2024.1439303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024] Open
Abstract
The early microbial colonization of human mucosal surfaces is essential for the development of the host immune system. Already during pregnancy, the unborn child is prepared for the postnatal influx of commensals and pathogens via maternal antibodies, and after birth this protection is continued with antibodies in breast milk. During this critical window of time, which extends from pregnancy to the first year of life, each encounter with a microorganism can influence children's immune response and can have a lifelong impact on their life. For example, there are numerous links between the development of allergies and an altered gut microbiome. However, the exact mechanisms behind microbial influences, also extending to how viruses influence host-microbe interactions, are incompletely understood. In this review, we address the impact of infants' first microbial encounters, how the immune system develops to interact with gut microbiota, and summarize how an altered immune response could be implied in allergies.
Collapse
Affiliation(s)
| | - Thomas Vogl
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Noble A, Paudyal B, Schwartz JC, Mwangi W, Munir D, Tchilian E, Hammond JA, Graham SP. Distinct effector functions mediated by Fc regions of bovine IgG subclasses and their interaction with Fc gamma receptors. Front Immunol 2023; 14:1286903. [PMID: 38077405 PMCID: PMC10702552 DOI: 10.3389/fimmu.2023.1286903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Cattle possess three IgG subclasses. However, the key immune functions, including complement and NK cell activation, and enhancement of phagocytosis, are not fully described for bovine IgG1, 2 and 3. We produced chimeric monoclonal antibodies (mAbs) consisting of a defined variable region linked to the constant regions of bovine IgG1, 2 and 3, and expressed His-tagged soluble recombinant bovine Fc gamma receptors (FcγRs) IA (CD64), IIA (CD32A), III (CD16) and Fcγ2R. Functional assays using bovinized mAbs were developed. IgG1 and IgG3, but not IgG2, activated complement-dependent cytotoxicity. Only IgG1 could activate cattle NK cells to mobilize CD107a after antigen crosslinking, a surrogate assay for antibody-dependent cell cytotoxicity. Both IgG1 and IgG2 could trigger monocyte-derived macrophages to phagocytose fluorescently labelled antigen-expressing target cells. IgG3 induced only weak antibody-dependent cellular phagocytosis (ADCP). By contrast, monocytes only exhibited strong ADCP when triggered by IgG2. IgG1 bound most strongly to recombinant FcγRs IA, IIA and III, with weaker binding by IgG3 and none by IgG2, which bound exclusively to Fcγ2R. Immune complexes containing IgG1, 2 and 3 bound differentially to leukocyte subsets, with IgG2 binding strongly to neutrophils and monocytes and all subclasses binding platelets. Differential expression of the FcγRs on leukocyte subsets was demonstrated by surface staining and/or RT-qPCR of sorted cells, e.g., Fcγ2R mRNA was expressed in monocytes/macrophages, neutrophils, and platelets, potentially explaining their strong interactions with IgG2, and FcγRIII was expressed on NK cells, presumably mediating IgG1-dependent NK cell activation. These data reveal differences in bovine IgG subclass functionality, which do not correspond to those described in humans, mice or pigs, which is relevant to the study of these IgG subclasses in vaccine and therapeutic antibody development.
Collapse
|
5
|
Dolatshahi S, Butler AL, Pou C, Henckel E, Bernhardsson AK, Gustafsson A, Bohlin K, Shin SA, Lauffenburger DA, Brodin P, Alter G. Selective transfer of maternal antibodies in preterm and fullterm children. Sci Rep 2022; 12:14937. [PMID: 36056073 PMCID: PMC9440225 DOI: 10.1038/s41598-022-18973-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 08/23/2022] [Indexed: 01/11/2023] Open
Abstract
Preterm newborns are more likely to suffer from infectious diseases at birth compared to children delivered at term. Whether this is due to compromised cellular, humoral, or organ-specific development remains unclear. To begin to define whether maternal-fetal antibody transfer profiles differ across preterm (PT) and fullterm (FT) infants, the overall quantity and functional quality of an array of 24 vaccine-, endemic pathogen-, and common antigen-specific antibodies were assessed across a cohort of 11 PT and 12 term-delivered maternal:infant pairs from birth through week 12. While total IgG levels to influenza, pneumo, measles, rubella, EBV, and RSV were higher in FT newborns, selective Fc-receptor binding antibodies was noted in PT newborns. In fact, near equivalent antibody-effector functions were observed across PT and FT infants, despite significant quantitative differences in transferred antibody levels. Moreover, temporal transfer analysis revealed the selective early transfer of FcRn, FcγR2, and FcγR3 binding antibodies, pointing to differential placental sieving mechanisms across gestation. These data point to selectivity in placental transfer at distinct gestational ages, to ensure that children are endowed with the most robust humoral immunity even if born preterm.
Collapse
Affiliation(s)
- Sepideh Dolatshahi
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, USA
| | | | - Christian Pou
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ewa Henckel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Anna Karin Bernhardsson
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Anna Gustafsson
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Kajsa Bohlin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Sally A Shin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering and Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
- Karolinska University Hospital, Stockholm, Sweden.
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
6
|
Dhande JR, Bagul RD, Thakar MR. HIV-gp140-Specific Antibodies Generated From Indian Long-Term Non-Progressors Mediate Potent ADCC Activity and Effectively Lyse Reactivated HIV Reservoir. Front Immunol 2022; 13:844610. [PMID: 35309295 PMCID: PMC8924355 DOI: 10.3389/fimmu.2022.844610] [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: 12/28/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Strategies to reduce the human immunodeficiency virus (HIV) reservoir are urgently required. The antibody-dependent cellular cytotoxicity (ADCC)-mediating anti-HIV antibodies have shown an association with HIV control. We assessed if such antibodies can be generated in vitro and whether the generated antibodies can facilitate the reduction of reactivated HIV reservoir. We isolated HIV-1-gp140-specific memory B cells from HIV-1-infected long-term non-progressors (LTNPs) with or without plasma ADCC and cultured them to generate anti-HIV antibodies. The ability of the generated antibodies to mediate ADCC and facilitate NK cell-mediated lysis of reactivated HIV reservoir was assessed by the rapid fluorometric antibody-dependent cellular cytotoxicity assay and a flow-based novel latency reduction assay, respectively. All LTNPs showed the presence of gp140-specific memory B cells [median: 0.79% (0.54%–1.225%)], which were successfully differentiated into plasma cells [median 72.0% (68.7–82.2%)] in an in-vitro culture and secreted antibodies [median OD: 0.253 (0.205–0.274)]. The HIV-gp140-specific antibodies were generated from 11/13 LTNPs irrespective of their plasma ADCC status. The generated antibodies from LTNPs with plasma ADCC showed higher ADCC potency (median: 37.6%, IQR: 32.95%–51%) and higher reduction in reactivated HIV reservoir (median: 62.5%, IQR: 58.71%–64.92%) as compared with the antibodies generated from LTNPs without plasma ADCC (ADCC: median: 8.85%, IQR: 8%–9.7%; and % p24 reduction median: 13.84, IQR: 9.863%–17.81%). The potency of these antibodies to reduce latent reservoir was two-fold higher than the respective plasma ADCC. The study showed that the potent ADCC-mediating antibodies could be generated from memory B cells of the LTNPs with plasma ADCC activity. These antibodies also showed potent ability to facilitate NK cell-mediated lysis of reactivated HIV reservoirs. It also indicated that memory B cells from individuals with plasma ADCC activity should be preferentially used for such antibody generation. The important role of these antibodies in the reduction of latent reservoirs needs to be further evaluated as a useful strategy to obtain a functional cure for HIV infection.
Collapse
|
7
|
Foss S, Jonsson A, Bottermann M, Watkinson R, Lode HE, McAdam MB, Michaelsen TE, Sandlie I, James LC, Andersen JT. Potent TRIM21 and complement-dependent intracellular antiviral immunity requires the IgG3 hinge. Sci Immunol 2022; 7:eabj1640. [PMID: 35486676 PMCID: PMC7614286 DOI: 10.1126/sciimmunol.abj1640] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Humans have four IgG antibody subclasses that selectively or differentially engage immune effector molecules to protect against infections. Although IgG1 has been studied in detail and is the subclass of most approved antibody therapeutics, increasing evidence indicates that IgG3 is associated with enhanced protection against pathogens. Here, we report that IgG3 has superior capacity to mediate intracellular antiviral immunity compared with the other subclasses due to its uniquely extended and flexible hinge region, which facilitates improved recruitment of the cytosolic Fc receptor TRIM21, independently of Fc binding affinity. TRIM21 may also synergize with complement C1/C4-mediated lysosomal degradation via capsid inactivation. We demonstrate that this process is potentiated by IgG3 in a hinge-dependent manner. Our findings reveal differences in how the four IgG subclasses mediate intracellular immunity, knowledge that may guide IgG subclass selection and engineering of antiviral antibodies for prophylaxis and therapy.
Collapse
Affiliation(s)
- Stian Foss
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, PO Box 4956, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| | - Alexandra Jonsson
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, PO Box 4956, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| | - Maria Bottermann
- Protein and Nucleic Acid Chemistry Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2-0QH, United Kingdom
| | - Ruth Watkinson
- Protein and Nucleic Acid Chemistry Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2-0QH, United Kingdom
| | - Heidrun E. Lode
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, PO Box 4956, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| | - Martin B. McAdam
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, PO Box 4956, N-0424 Oslo, Norway
| | - Terje E. Michaelsen
- Department of Chemical Pharmacy, School of Pharmacy, University of Oslo, N-0371, Oslo, Norway
- Infection Immunology, Norwegian Institute of Public Health, N-0213, Oslo, Norway
| | - Inger Sandlie
- Centre for Immune Regulation (CIR) and Department of Biosciences, University of Oslo, N-0371 Oslo, Norway
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, PO Box 4956, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| | - Leo C. James
- Protein and Nucleic Acid Chemistry Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, CB2-0QH, United Kingdom
| | - Jan Terje Andersen
- CIR and Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, PO Box 4956, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| |
Collapse
|
8
|
Evolution of antibodies to native trimeric envelope and their Fc dependent functions in untreated and treated primary HIV infection. J Virol 2021; 95:e0162521. [PMID: 34586863 DOI: 10.1128/jvi.01625-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
People living with HIV (PLWH) develop both anti-Envelope-specific antibodies, which bind the closed trimeric HIV Envelope present on infected cells and anti-gp120-specific antibodies, which bind gp120 monomers shed by infected cells and taken up by CD4 on uninfected bystander cells. Both antibodies have an Fc portion that binds to Fc Receptors on several types of innate immune cells and stimulates them to develop anti-viral functions. Among these Fc dependent functions (FcDFs) are antibody dependent (AD) cellular cytotoxicity (ADCC), AD cellular trogocytosis (ADCT) and AD phagocytosis (ADCP). Here, we assessed the evolution of total immunoglobulin G (IgG), anti-gp120 and anti-Envelope IgG antibodies and their FcDFs in plasma samples from anti-retroviral therapy (ART) naïve subjects during early HIV infection (28-194 days post infection [DPI]). We found that both the concentrations and FcDFs of anti-gp120 and anti-Envelope antibodies increased with time in ART-naïve PLWH. Although generated concurrently, anti-gp120-specific antibodies were 20.7-fold more abundant than anti-Envelpe-specific antibodies, both specificities being strongly correlated with each other and FcDFs. Among the FcDFs, only ADCP activity was inversely correlated with concurrent viral load. PLWH who started ART >90 DPI showed higher anti-Envelope-specific antibody levels, ADCT and ADCP activities than those starting ART <90 DPI. However, in longitudinally collected samples, ART initiation at >90 DPI was accompanied by a faster decline in anti-Envelope-specific antibody levels, which did not translate to a faster decline in FcDFs compared to those starting ART <90 DPI. IMPORTANCE Closed conformation Envelope is expressed on the surface of HIV-infected cells. Antibodies targeting this conformation and that support FcDFs have the potential to control HIV. This study tracks the timing of the appearance and evolution of antibodies to closed conformation Envelope, whose concentration increases over the first 6 mos of infection. Antiretroviral therapy (ART) initiation blunts further increases in the concentration of these antibodies and their and FcDFs. However, antibodies to open conformation Envelope also increase with DPI until ART initiation. These antibodies target uninfected bystander cells, which may contribute to loss of uninfected CD4 cells and pathogenicity. This manuscript presents, for the first time, the evolution of antibodies to closed conformation Envelope and their fate on-ART. This information may be useful in making decisions on the timing of ART initiation in early HIV infection.
Collapse
|
9
|
Doepker LE, Danon S, Harkins E, Ralph DK, Yaffe Z, Garrett ME, Dhar A, Wagner C, Stumpf MM, Arenz D, Williams JA, Jaoko W, Mandaliya K, Lee KK, Matsen FA, Overbaugh JM. Development of antibody-dependent cell cytotoxicity function in HIV-1 antibodies. eLife 2021; 10:e63444. [PMID: 33427196 PMCID: PMC7884072 DOI: 10.7554/elife.63444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/08/2021] [Indexed: 11/27/2022] Open
Abstract
A prerequisite for the design of an HIV vaccine that elicits protective antibodies is understanding the developmental pathways that result in desirable antibody features. The development of antibodies that mediate antibody-dependent cellular cytotoxicity (ADCC) is particularly relevant because such antibodies have been associated with HIV protection in humans. We reconstructed the developmental pathways of six human HIV-specific ADCC antibodies using longitudinal antibody sequencing data. Most of the inferred naive antibodies did not mediate detectable ADCC. Gain of antigen binding and ADCC function typically required mutations in complementarity determining regions of one or both chains. Enhancement of ADCC potency often required additional mutations in framework regions. Antigen binding affinity and ADCC activity were correlated, but affinity alone was not sufficient to predict ADCC potency. Thus, elicitation of broadly active ADCC antibodies may require mutations that enable high-affinity antigen recognition along with mutations that optimize factors contributing to functional ADCC activity.
Collapse
Affiliation(s)
- Laura E Doepker
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Sonja Danon
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Elias Harkins
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Duncan K Ralph
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Zak Yaffe
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Medical Scientist Training Program, University of Washington School of MedicineSeattleUnited States
| | - Meghan E Garrett
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Molecular and Cellular Biology Graduate Program, University of Washington and Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Amrit Dhar
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Department of Statistics, University of WashingtonSeattleUnited States
| | - Cassia Wagner
- Medical Scientist Training Program, University of Washington School of MedicineSeattleUnited States
| | - Megan M Stumpf
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Dana Arenz
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - James A Williams
- Department of Medicinal Chemistry, University of WashingtonSeattleUnited States
| | - Walter Jaoko
- Department of Medicinal Microbiology, University of NairobiNairobiKenya
| | - Kishor Mandaliya
- Coast Provincial General Hospital, Women’s Health ProjectMombasaKenya
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of WashingtonSeattleUnited States
| | - Frederick A Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| | - Julie M Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleUnited States
| |
Collapse
|
10
|
Chu TH, Patz EF, Ackerman ME. Coming together at the hinges: Therapeutic prospects of IgG3. MAbs 2021; 13:1882028. [PMID: 33602056 PMCID: PMC7899677 DOI: 10.1080/19420862.2021.1882028] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/08/2021] [Accepted: 01/22/2021] [Indexed: 01/22/2023] Open
Abstract
The human IgG3 subclass is conspicuously absent among the formats for approved monoclonal antibody therapies and Fc fusion protein biologics. Concern about the potential for rapid degradation, reduced plasma half-life, and increased immunogenicity due to marked variation in allotypes has apparently outweighed the potential advantages of IgG3, which include high affinity for activating Fcγ receptors, effective complement fixation, and a long hinge that appears better suited for low abundance targets. This review aims to highlight distinguishing features of IgG3 and to explore its functional role in the immune response. We present studies of natural immunity and recombinant antibody therapies that elucidate key contributions of IgG3 and discuss historical roadblocks that no longer remain clearly relevant. Collectively, this body of evidence motivates thoughtful reconsideration of the clinical advancement of this distinctive antibody subclass for treatment of human diseases. Abbreviations: ADCC - Antibody-Dependent Cell-mediated CytotoxicityADE - Antibody-dependent enhancementAID - Activation-Induced Cytidine DeaminaseCH - Constant HeavyCHF - Complement factor HCSR - Class Switch RecombinationEM - Electron MicroscopyFab - Fragment, antigen bindingFc - Fragment, crystallizableFcRn - Neonatal Fc ReceptorFcγR - Fc gamma ReceptorHIV - Human Immunodeficiency VirusIg - ImmunoglobulinIgH - Immunoglobulin Heavy chain geneNHP - Non-Human Primate.
Collapse
Affiliation(s)
- Thach H. Chu
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Edward F. Patz
- Department of Radiology and Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | | |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Rare patients naturally control HIV replication without antiretroviral therapy. Understanding the mechanisms implicated in natural HIV control will inform the development of immunotherapies against HIV. Elite controllers are known for developing efficient antiviral T-cell responses, but recent findings suggest that antibody responses also play a significant role in HIV control. We review the key studies that uncovered a potent memory B-cell response and highly functional anti-HIV antibodies in elite controllers, and explore the mechanisms that may account for the distinct properties of their humoral response. RECENT FINDINGS Elite controllers maintain a large HIV-specific memory B-cell pool that is sustained by efficient T follicular helper function. Neutralizing antibody rarely show high titers in controllers, but seem capable, at least in certain cases, of neutralizing contemporaneous viral strains. In addition, elite controllers display a unique HIV-specific antibody profile in terms of isotype, antigen specificity, and glycosylation pattern, resulting in polyfunctional antibody effector functions that may promote infected cell lysis and prime effectors of the antiviral immune response. SUMMARY Lessons from elite controller studies argue for the importance of integrating the many parameters defining a polyfunctional antibody response when evaluating candidate vaccines and immunotherapeutic approaches directed at HIV.
Collapse
|
12
|
Systems serology for decoding infection and vaccine-induced antibody responses to HIV-1. Curr Opin HIV AIDS 2020; 14:253-264. [PMID: 31033729 DOI: 10.1097/coh.0000000000000558] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Experimental and analytical advances have enabled systematic, high-resolution studies of humoral immune responses, and are beginning to define mechanisms of immunity to HIV. RECENT FINDINGS High-throughput, information-rich experimental and analytical methods, whether genomic, proteomic, or transcriptomic, have firmly established their value across a diversity of fields. Consideration of these tools as trawlers in 'fishing expeditions' has faded as 'data-driven discovery' has come to be valued as an irreplaceable means to develop fundamental understanding of biological systems. Collectively, studies of HIV-1 infection and vaccination including functional, biophysical, and biochemical humoral profiling approaches have provided insights into the phenotypic characteristics of individual and pools of antibodies. Relating these measures to clinical status, protection/efficacy outcomes, and cellular profiling data using machine learning has offered the possibility of identifying unanticipated mechanisms of action and gaining insights into fundamental immunological processes that might otherwise be difficult to decipher. SUMMARY Recent evidence establishes that systematic data collection and application of machine learning approaches can identify humoral immune correlates that are generalizable across distinct HIV-1 immunogens and vaccine regimens and translatable between model organisms and the clinic. These outcomes provide a strong rationale supporting the utility and further expansion of these approaches both in support of vaccine development and more broadly in defining mechanisms of immunity.
Collapse
|
13
|
Chu TH, Crowley AR, Backes I, Chang C, Tay M, Broge T, Tuyishime M, Ferrari G, Seaman MS, Richardson SI, Tomaras GD, Alter G, Leib D, Ackerman ME. Hinge length contributes to the phagocytic activity of HIV-specific IgG1 and IgG3 antibodies. PLoS Pathog 2020; 16:e1008083. [PMID: 32092122 PMCID: PMC7058349 DOI: 10.1371/journal.ppat.1008083] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 03/05/2020] [Accepted: 09/16/2019] [Indexed: 12/13/2022] Open
Abstract
Antibody functions such as neutralization require recognition of antigen by the Fab region, while effector functions are additionally mediated by interactions of the Fc region with soluble factors and cellular receptors. The efficacy of individual antibodies varies based on Fab domain characteristics, such as affinity for antigen and epitope-specificity, and on Fc domain characteristics that include isotype, subclass, and glycosylation profile. Here, a series of HIV-specific antibody subclass and hinge variants were constructed and tested to define those properties associated with differential effector function. In the context of the broadly neutralizing CD4 binding site-specific antibody VRC01 and the variable loop (V3) binding antibody 447-52D, hinge truncation and extension had a considerable impact on the magnitude of phagocytic activity of both IgG1 and IgG3 subclasses. The improvement in phagocytic potency of antibodies with extended hinges could not be attributed to changes in either intrinsic antigen or antibody receptor affinity. This effect was specific to phagocytosis and was generalizable to different phagocytes, at different effector cell to target ratios, for target particles of different size and composition, and occurred across a range of antibody concentrations. Antibody dependent cellular cytotoxicity and neutralization were generally independent of hinge length, and complement deposition displayed variable local optima. In vivo stability testing showed that IgG molecules with altered hinges can exhibit similar biodistribution and pharmacokinetic profiles as IgG1. Overall, these results suggest that when high phagocytic activity is desirable, therapeutic antibodies may benefit from being formatted as human IgG3 or engineered IgG1 forms with elongated hinges.
Collapse
Affiliation(s)
- Thach H. Chu
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Andrew R. Crowley
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Iara Backes
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Cheryl Chang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Matthew Tay
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Thomas Broge
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Marina Tuyishime
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael S. Seaman
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Simone I. Richardson
- Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, Gauteng, South Africa
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - David Leib
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
- The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| |
Collapse
|
14
|
McLean MR, Lu LL, Kent SJ, Chung AW. An Inflammatory Story: Antibodies in Tuberculosis Comorbidities. Front Immunol 2019; 10:2846. [PMID: 31921122 PMCID: PMC6913197 DOI: 10.3389/fimmu.2019.02846] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) resides in a quarter of the world's population and is the causative agent for tuberculosis (TB), the most common infectious reason of death in humans today. Although cellular immunity has been firmly established in the control of Mtb, there is growing evidence that antibodies may also modulate the infection. More specifically, certain antibody features are associated with inflammation and are divergent in different states of human infection and disease. Importantly, TB impacts not just the healthy but also those with chronic conditions. While HIV represents the quintessential comorbid condition for TB, recent epidemiological evidence shows that additional chronic conditions such as diabetes and kidney disease are rising. In fact, the prevalence of diabetes as a comorbid TB condition is now higher than that of HIV. These chronic diseases are themselves independently associated with pro-inflammatory immune states that encompass antibody profiles. This review discusses isotypes, subclasses, post-translational modifications and Fc-mediated functions of antibodies in TB infection and in the comorbid chronic conditions of HIV, diabetes, and kidney diseases. We propose that inflammatory antibody profiles, which are a marker of active TB, may be an important biomarker for detection of TB disease progression within comorbid individuals. We highlight the need for future studies to determine which inflammatory antibody profiles are the consequences of comorbidities and which may potentially contribute to TB reactivation.
Collapse
Affiliation(s)
- Milla R McLean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Lenette L Lu
- Division of Infectious Disease and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Infectious Diseases Department, Melbourne Sexual Health Centre, Alfred Health, Central Clinical School, Monash University, Brisbane, VIC, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, SA, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
15
|
Richardson SI, Lambson BE, Crowley AR, Bashirova A, Scheepers C, Garrett N, Abdool Karim S, Mkhize NN, Carrington M, Ackerman ME, Moore PL, Morris L. IgG3 enhances neutralization potency and Fc effector function of an HIV V2-specific broadly neutralizing antibody. PLoS Pathog 2019; 15:e1008064. [PMID: 31841557 PMCID: PMC6936867 DOI: 10.1371/journal.ppat.1008064] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/30/2019] [Accepted: 09/02/2019] [Indexed: 11/19/2022] Open
Abstract
Broadly neutralizing antibodies (bNAbs) protect against HIV infection in non-human primates and their efficacy may be enhanced through interaction with Fc receptors on immune cells. Antibody isotype is a modulator of this binding with the IgG3 subclass mediating potent Fc effector function and is associated with HIV vaccine efficacy and HIV control. BNAb functions are typically assessed independently of the constant region with which they are naturally expressed. To examine the role of natural isotype in the context of a bNAb lineage we studied CAP256, an HIV-infected individual that mounted a potent V2-specific bNAb response. CAP256 expressed persistently high levels of plasma IgG3 which we found mediated both broad neutralizing activity and potent Fc function. Sequencing of germline DNA and the constant regions of V2-directed bNAbs from this donor revealed the expression of a novel IGHG3 allele as well as IGHG3*17, an allele that produces IgG3 antibodies with increased plasma half-life. Both allelic variants were used to generate CAP256-VRC26.25 and CAP256-VRC26.29 IgG3 bNAbs and these were compared to IgG1 versions. IgG3 variants were shown to have significantly higher phagocytosis and trogocytosis compared to IgG1 versions, which corresponded to increased affinity for FcγRIIa. Neutralization potency was also significantly higher for IgG3 bNAbs, particularly against viruses lacking the N160 glycan. By exchanging hinge regions between subclass variants, we showed that hinge length modulated both neutralization potency and Fc function. This study showed that co-operation between the variable and natural IgG3 constant regions enhanced the polyfunctionality of antibodies, indicating the value of leveraging genetic variation which could be exploited for passive immunity.
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
| | - Bronwen E. Lambson
- 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
| | - Andrew R. Crowley
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Arman Bashirova
- Ragon Institute of Massachusetts General Hospital, MIT, and Harvard University, Boston, Massachusetts, United States of America
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Cathrine Scheepers
- 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, Congella, KwaZulu-Natal, South Africa
- Department of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
| | - Salim Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Congella, KwaZulu-Natal, South Africa
- Department of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
- Department of Epidemiology, Columbia University, New York, NY, United States of America
| | - 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
| | - Mary Carrington
- Ragon Institute of Massachusetts General Hospital, MIT, and Harvard University, Boston, Massachusetts, United States of America
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - 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, Congella, KwaZulu-Natal, South Africa
| | - 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, Congella, KwaZulu-Natal, South Africa
| |
Collapse
|
16
|
Foss S, Bottermann M, Jonsson A, Sandlie I, James LC, Andersen JT. TRIM21-From Intracellular Immunity to Therapy. Front Immunol 2019; 10:2049. [PMID: 31555278 PMCID: PMC6722209 DOI: 10.3389/fimmu.2019.02049] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022] Open
Abstract
Tripartite motif containing-21 (TRIM21) is a cytosolic ubiquitin ligase and antibody receptor that provides a last line of defense against invading viruses. It does so by acting as a sensor that intercepts antibody-coated viruses that have evaded extracellular neutralization and breached the cell membrane. Upon engagement of the Fc of antibodies bound to viruses, TRIM21 triggers a coordinated effector and signaling response that prevents viral replication while at the same time inducing an anti-viral cellular state. This dual effector function is tightly regulated by auto-ubiquitination and phosphorylation. Therapeutically, TRIM21 has been shown to be detrimental in adenovirus based gene therapy, while it may be favorably utilized to prevent tau aggregation in neurodegenerative disorders. In addition, TRIM21 may synergize with the complement system to block viral replication as well as transgene expression. TRIM21 can also be utilized as a research tool to deplete specific proteins in cells and zebrafish embryos. Here, we review our current biological understanding of TRIM21 in light of its versatile functions.
Collapse
Affiliation(s)
- Stian Foss
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Maria Bottermann
- Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Medical Research Council, Cambridge, United Kingdom
| | - Alexandra Jonsson
- Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway.,Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Medical Research Council, Cambridge, United Kingdom
| | - Inger Sandlie
- Department of Biosciences, Centre for Immune Regulation, University of Oslo, Oslo, Norway.,Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Leo C James
- Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Medical Research Council, Cambridge, United Kingdom
| | - Jan Terje Andersen
- Department of Immunology, Centre for Immune Regulation, Rikshospitalet, Oslo University Hospital and University of Oslo, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Crowley AR, Ackerman ME. Mind the Gap: How Interspecies Variability in IgG and Its Receptors May Complicate Comparisons of Human and Non-human Primate Effector Function. Front Immunol 2019; 10:697. [PMID: 31024542 PMCID: PMC6463756 DOI: 10.3389/fimmu.2019.00697] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/13/2019] [Indexed: 01/08/2023] Open
Abstract
The field of HIV research relies heavily on non-human primates, particularly the members of the macaque genus, as models for the evaluation of candidate vaccines and monoclonal antibodies. A growing body of research suggests that successful protection of humans will not solely rely on the neutralization activity of an antibody's antigen binding fragment. Rather, immunological effector functions prompted by the interaction of the immunoglobulin G constant region and its cognate Fc receptors help contribute to favorable outcomes. Inherent differences in the sequences, expression, and activities of human and non-human primate antibody receptors and immunoglobulins have the potential to produce disparate results in the observations made in studies conducted in differing species. Having a more complete understanding of these differences, however, should permit the more fluent translation of observations between model organisms and the clinic. Here we present a guide to such translations that encompasses not only what is presently known regarding the affinity of the receptor-ligand interactions but also the influence of expression patterns and allelic variation, with a focus on insights gained from use of this model in HIV vaccines and passive antibody therapy and treatment.
Collapse
Affiliation(s)
- Andrew R. Crowley
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH, United States
| | - Margaret E. Ackerman
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The design of an HIV vaccine remains an elusive but top priority. Data from the non-human primate model and the first moderately protective HIV vaccine trial (RV144) point to a role for qualitative changes in humoral immune functions in protection from infection. Here, we review the current understanding of the antibody response throughout HIV infection, the known correlates of protection, and current strategies to manipulate antibodies to put an end to the epidemic. RECENT FINDINGS Recent studies point to innate immune-recruiting antibody function in preventing infection as well as controlling viremia following infection. These data have begun to inform next-generation design of HIV vaccines and antibody therapies by uncovering new viral targets and antibody architectures to improve potency and breadth. Emerging data illustrate a role for innate immune recruiting-antibodies in conferring protection against HIV infection as well as promoting viral control and clearance, offering an unprecedented opportunity to modulate and improve antibody function to fight HIV more effectively.
Collapse
Affiliation(s)
- Audrey L. Butler
- The Ragon Institute of MGH, MIT, and Harvard, 400 Technology Square, Cambridge, MA 02139 USA
| | - Stephanie Fischinger
- The Ragon Institute of MGH, MIT, and Harvard, 400 Technology Square, Cambridge, MA 02139 USA
| | - Galit Alter
- The Ragon Institute of MGH, MIT, and Harvard, 400 Technology Square, Cambridge, MA 02139 USA
| |
Collapse
|
19
|
Damelang T, Rogerson SJ, Kent SJ, Chung AW. Role of IgG3 in Infectious Diseases. Trends Immunol 2019; 40:197-211. [PMID: 30745265 DOI: 10.1016/j.it.2019.01.005] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022]
Abstract
IgG3 comprises only a minor fraction of IgG and has remained relatively understudied until recent years. Key physiochemical characteristics of IgG3 include an elongated hinge region, greater molecular flexibility, extensive polymorphisms, and additional glycosylation sites not present on other IgG subclasses. These characteristics make IgG3 a uniquely potent immunoglobulin, with the potential for triggering effector functions including complement activation, antibody (Ab)-mediated phagocytosis, or Ab-mediated cellular cytotoxicity (ADCC). Recent studies underscore the importance of IgG3 effector functions against a range of pathogens and have provided approaches to overcome IgG3-associated limitations, such as allotype-dependent short Ab half-life, and excessive proinflammatory activation. Understanding the molecular and functional properties of IgG3 may facilitate the development of improved Ab-based immunotherapies and vaccines against infectious diseases.
Collapse
Affiliation(s)
- Timon Damelang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Medicine, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
20
|
Lopez E, Shattock RJ, Kent SJ, Chung AW. The Multifaceted Nature of Immunoglobulin A and Its Complex Role in HIV. AIDS Res Hum Retroviruses 2018; 34:727-738. [PMID: 30056749 DOI: 10.1089/aid.2018.0099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IgA is the most abundant immunoglobulin in mucosal secretions, and understanding the role of IgA in both protection from HIV acquisition and modulation of HIV disease progression is a field of considerable controversy and renewed research interest. Analysis of the RV144 clinical trial associated plasma HIV envelope-specific monomeric IgA from vaccines with reduced vaccine efficacy. The RV144 trial, however, only assessed for plasma IgA, which was not further subclassed, and the role of mucosal IgA was not addressed as mucosal samples were not collected. On the other hand, several studies have detected envelope-specific IgA in mucosal secretions of highly exposed persistently seronegative cohorts, while recent macaque simian-HIV passive immunization studies have suggested a potentially protective role for mucosal IgA. It is well established that total IgA in serum appears to correlate with HIV disease progression. In contrast, a selective deficit of anti-HIV IgA responses in HIV infection is apparent, with a number of recent studies beginning to elucidate the mechanisms behind these dysfunctional IgA responses. In this review, we highlight the dichotomy that exists in the literature as to whether anti-HIV IgA is protective or harmful to the host. Herein, we emphasize the importance of distinguishing between monomeric, multimeric, and isoforms of IgA and review what is known about the complex and diverse interactions of various molecular forms of IgA with HIV in both the systemic circulation and mucosal compartments.
Collapse
Affiliation(s)
- Ester Lopez
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Robin J. Shattock
- Mucosal Infection and Immunity Group, Department of Medicine, Imperial College London, London, United Kingdom
| | - Stephen J. Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
- Infectious Diseases Department, Melbourne Sexual Health Centre, Alfred Health, Central Clinical School, Monash University, Melbourne, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Melbourne, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| |
Collapse
|
21
|
IgG3 regulates tissue-like memory B cells in HIV-infected individuals. Nat Immunol 2018; 19:1001-1012. [PMID: 30104633 DOI: 10.1038/s41590-018-0180-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 06/28/2018] [Indexed: 01/10/2023]
Abstract
Immunoglobulin G3 (IgG3) has an uncertain role in the response to infection with and vaccination against human immunodeficiency virus (HIV). Here we describe a regulatory role for IgG3 in dampening the immune system-activating effects of chronic HIV viremia on B cells. Secreted IgG3 was bound to IgM-expressing B cells in vivo in HIV-infected chronically viremic individuals but not in early-viremic or aviremic individuals. Tissue-like memory (TLM) B cells, a population expanded by persistent HIV viremia, bound large amounts of IgG3. IgG3 induced clustering of B cell antigen receptors (BCRs) on the IgM+ B cells, which was mediated by direct interactions between soluble IgG3 and membrane IgM of the BCR (IgM-BCR). The inhibitory IgG receptor CD32b (FcγRIIb), complement component C1q and inflammatory biomarker CRP contributed to the binding of secreted IgG3 onto IgM-expressing B cells of HIV-infected individuals. Notably, IgG3-bound TLM B cells were refractory to IgM-BCR stimulation, thus demonstrating that IgG3 can regulate B cells during chronic activation of the immune system.
Collapse
|
22
|
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
|
23
|
Temporal variation in HIV-specific IgG subclass antibodies during acute infection differentiates spontaneous controllers from chronic progressors. AIDS 2018; 32:443-450. [PMID: 29239894 DOI: 10.1097/qad.0000000000001716] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Given the emerging appreciation for the role of antibody-dependent effector functions and IgG subclass distribution among spontaneous controllers of HIV, we sought to determine whether antibody-associated features diverged in early HIV infection between patients who ultimately became controllers versus those who became progressors. METHODS IgG was purified from plasma from nine acutely infected patients who subsequently controlled HIV spontaneously (controllers) and 10 acutely infected individuals who did not control viremia (progressors). Antibody profiles were compared at weeks 4, 12, 24 and 48 postinfection. Levels of clade B gp120-specific, gp140-specific and gp41-specific IgG antibody subclasses were measured. In addition, gp120-specific antibody-dependent cellular phagocytosis, rapid fluorescent antibody-dependent cellular cytotoxicity and antibody-dependent cellular viral inhibition were all assessed. RESULTS Although no single antibody-related measurement was significantly associated with long-term HIV control, combinations of antibody-associated variables were able to accurately differentiate controllers and progressors. In contrast to controllers, progressors showed greater dynamic changes in gp120-specific subclass selection profiles, with increasing levels of Env-specific IgG2 antibodies and losses in Env-specific IgG3 antibodies. Moreover, progressors, but not controllers, lost antibody-dependent cellular viral inhibition function over time. Together, these results highlight changes in IgG subclass selection profiles in progressive, but not controlled, HIV infection. CONCLUSION This study suggests that the temporal variation and maintenance of Env-specific IgG subclasses during acute HIV infection are predictive of eventual disease control. The maintenance of gp120-specific and gp140-specific IgG3 may contribute to control of disease in spontaneous controllers. Thus, strategies to induce stable IgG3 responses may preserve control of the viral reservoir.
Collapse
|
24
|
Ruiz MJ, Salido J, Abusamra L, Ghiglione Y, Cevallos C, Damilano G, Rodriguez AM, Trifone C, Laufer N, Giavedoni LD, Sued O, Salomón H, Gherardi MM, Turk G. Evaluation of Different Parameters of Humoral and Cellular Immune Responses in HIV Serodiscordant Heterosexual Couples: Humoral Response Potentially Implicated in Modulating Transmission Rates. EBioMedicine 2017; 26:25-37. [PMID: 29129698 PMCID: PMC5832641 DOI: 10.1016/j.ebiom.2017.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 02/05/2023] Open
Abstract
As the HIV/AIDS pandemic still progresses, understanding the mechanisms governing viral transmission as well as protection from HIV acquisition is fundamental. In this context, cohorts of HIV serodiscordant heterosexual couples (SDC) represent a unique tool. The present study was aimed to evaluate specific parameters of innate, cellular and humoral immune responses in SDC. Specifically, plasma levels of cytokines and chemokines, HIV-specific T-cell responses, gp120-specific IgG and IgA antibodies, and HIV-specific antibody-dependent cellular cytotoxicity (ADCC) activity were assessed in nine HIV-exposed seronegative individuals (ESN) and their corresponding HIV seropositive partners (HIV+-P), in eighteen chronically infected HIV subjects (C), nine chronically infected subjects known to be HIV transmitters (CT) and ten healthy HIV− donors (HD). Very low magnitude HIV-specific cellular responses were found in two out of six ESN. Interestingly, HIV+-P had the highest ADCC magnitude, the lowest IgA levels and the highest IgG/IgA ratio, all compared to CT. Positive correlations between CD4+ T-cell counts and both IgG/IgA ratios and %ADCC killing uniquely distinguished HIV+-P. Additionally, evidence of IgA interference with ADCC responses from HIV+-P and CT is provided. These data suggest for the first time a potential role of ADCC and/or gp120-specific IgG/IgA balance in modulating heterosexual transmission. In sum, this study provides key information to understand the host factors that influence viral transmission, which should be considered in both the development of prophylactic vaccines and novel immunotherapies for HIV-1 infection. The evaluation of different immune parameters in HIV serodiscordant couples helped identify factors shaping transmission. Innate and cellular immune responses were apparently not involved in this scenario. HIV-specific ADCC, IgA titer and IgG/IgA balance were identified as factors involved in modulating viral transmission.
The existence of individuals that remain HIV negative despite being repeatedly exposed to the virus has long been described. To date, only homozygosis for a 32-base pair deletion in the ccr5 gene has been consistently shown to be a determinant of HIV resistance. Still, subjects bearing the WT ccr5 gene have also been described as resistant or less susceptible to HIV. Thus, other mechanisms must be involved in this phenomenon. The results presented here postulate ADCC and IgG/IgA ratio as potential mechanisms involved in modulating HIV transmission in the context of serodiscordant couples and inspire further investigations.
Collapse
Affiliation(s)
- María Julia Ruiz
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Jimena Salido
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | | | - Yanina Ghiglione
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Cintia Cevallos
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Gabriel Damilano
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Ana María Rodriguez
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - César Trifone
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Natalia Laufer
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina; Hospital Juan A. Fernández, Unidad Enfermedades Infecciosas, Buenos Aires, Argentina
| | - Luis D Giavedoni
- Department of Virology and Immunology, Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina; Hospital Juan A. Fernández, Unidad Enfermedades Infecciosas, Buenos Aires, Argentina
| | - Horacio Salomón
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - María Magdalena Gherardi
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina
| | - Gabriela Turk
- CONICET- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Buenos Aires, Argentina.
| |
Collapse
|
25
|
Vanderven HA, Liu L, Ana-Sosa-Batiz F, Nguyen TH, Wan Y, Wines B, Hogarth PM, Tilmanis D, Reynaldi A, Parsons MS, Hurt AC, Davenport MP, Kotsimbos T, Cheng AC, Kedzierska K, Zhang X, Xu J, Kent SJ. Fc functional antibodies in humans with severe H7N9 and seasonal influenza. JCI Insight 2017; 2:92750. [PMID: 28679958 DOI: 10.1172/jci.insight.92750] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/19/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Both seasonal and novel avian influenza viruses can result in severe infections requiring hospitalization. Anti-influenza antibodies (Abs) with Fc-mediated effector functions, such as Ab-dependent cellular cytotoxicity (ADCC), are of growing interest in control of influenza but have not previously been studied during severe human infections. As such, the objective of this study was to examine Fc-mediated Ab functions in humans hospitalized with influenza infection. METHODS Serum Ab response was studied in subjects hospitalized with either pandemic H7N9 avian influenza virus in China (n = 18) or circulating seasonal influenza viruses in Melbourne, Australia (n = 16). Recombinant soluble Fc receptor dimer ELISAs, natural killer (NK) cell activation assays, and Ab-dependent killing assays with influenza-infected target cells were used to assess the Fc functionality of anti-influenza hemagglutinin (HA) Abs during severe human influenza infection. RESULTS We found that the peak generation of Fc functional HA Abs preceded that of neutralizing Abs for both severe H7N9 and seasonal influenza infections. Subjects who succumbed to complications of H7N9 infection demonstrated reduced HA-specific Fc receptor-binding Abs (in magnitude and breadth) immediately prior to death compared with those who survived. Subjects who recovered from H7N9 and severe seasonal influenza infections demonstrated increased Fc receptor-binding Abs not only against the homologous infecting strain but against HAs from different influenza A subtypes. CONCLUSION Collectively, survivors of severe influenza infection rapidly generate a functional Ab response capable of mediating ADCC against divergent influenza viruses. Broadly binding HA Abs with Fc-mediated functions may be a useful component of protective immunity to severe influenza infection. FUNDING The National Health and Medical Research Council ([NHMRC] grants 1023294, 1041832, and 1071916), the Australian Department of Health, and the joint University of Melbourne/Fudan University International Research and Research Training Fund provided funding for this study.
Collapse
Affiliation(s)
- Hillary A Vanderven
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Lu Liu
- Shanghai Public Health Clinical Centre (SPHCC) and Institute of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of the Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fernanda Ana-Sosa-Batiz
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Thi Ho Nguyen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Yanmin Wan
- Shanghai Public Health Clinical Centre (SPHCC) and Institute of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of the Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bruce Wines
- Burnet Institute, Melbourne, Victoria, Australia
| | | | - Danielle Tilmanis
- WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Arnold Reynaldi
- Infection Analytics Program, Kirby Institute for Infection and Immunity, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Matthew S Parsons
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Aeron C Hurt
- WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Miles P Davenport
- Infection Analytics Program, Kirby Institute for Infection and Immunity, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Tom Kotsimbos
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Allen C Cheng
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health and School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Centre (SPHCC) and Institute of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of the Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Centre (SPHCC) and Institute of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of the Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia.,Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
26
|
French MA, Tjiam MC, Abudulai LN, Fernandez S. Antiviral Functions of Human Immunodeficiency Virus Type 1 (HIV-1)-Specific IgG Antibodies: Effects of Antiretroviral Therapy and Implications for Therapeutic HIV-1 Vaccine Design. Front Immunol 2017; 8:780. [PMID: 28725225 PMCID: PMC5495868 DOI: 10.3389/fimmu.2017.00780] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/19/2017] [Indexed: 12/24/2022] Open
Abstract
Contemporary antiretroviral therapy (ART) is effective and tolerable for long periods of time but cannot eradicate human immunodeficiency virus type 1 (HIV-1) infection by either elimination of viral reservoirs or enhancement of HIV-1-specific immune responses. Boosting "protective" HIV-1-specific immune responses by active or passive immunization will therefore be necessary to control or eradicate HIV-1 infection and is currently the topic of intense investigation. Recently reported studies conducted in HIV patients and non-human primate (NHP) models of HIV-1 infection suggest that HIV-1-specific IgG antibody responses may contribute to the control of HIV-1 infection. However, production of IgG antibodies with virus neutralizing activity by vaccination remains problematic and while vaccine-induced natural killer cell-activating IgG antibodies have been shown to prevent the acquisition of HIV-1 infection, they may not be sufficient to control or eradicate established HIV-1 infection. It is, therefore, important to consider other functional characteristics of IgG antibody responses. IgG antibodies to viruses also mediate opsonophagocytic antibody responses against virions and capsids that enhance the function of phagocytic cells playing critical roles in antiviral immune responses, particularly conventional dendritic cells and plasmacytoid dendritic cells. Emerging evidence suggests that these antibody functions might contribute to the control of HIV-1 infection. In addition, IgG antibodies contribute to the intracellular degradation of viruses via binding to the cytosolic fragment crystallizable (Fc) receptor tripartite motif containing-21 (TRIM21). The functional activity of an IgG antibody response is influenced by the IgG subclass content, which affects binding to antigens and to Fcγ receptors on phagocytic cells and to TRIM21. The IgG subclass content and avidity of IgG antibodies is determined by germinal center (GC) reactions in follicles of lymphoid tissue. As HIV-1 infects cells in GCs and induces GC dysfunction, which may persist during ART, strategies for boosting HIV-1-specific IgG antibody responses should include early commencement of ART and possibly the use of particular antiretroviral drugs to optimize drug levels in lymphoid follicles. Finally, enhancing particular functions of HIV-1-specific IgG antibody responses by using adjuvants or cytokines to modulate the IgG subclass content of the antibody response might be investigated in NHP models of HIV-1 infection and during trials of therapeutic vaccines in HIV patients.
Collapse
Affiliation(s)
- Martyn A. French
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- Medical School, University of Western Australia, Perth, WA, Australia
- Department of Clinical Immunology, Royal Perth Hospital and PathWest Laboratory Medicine, Perth, WA, Australia
| | - M. Christian Tjiam
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Laila N. Abudulai
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Sonia Fernandez
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
27
|
Alvarez RA, Maestre AM, Law K, Durham ND, Barria MI, Ishii-Watabe A, Tada M, Kapoor M, Hotta MT, Rodriguez-Caprio G, Fierer DS, Fernandez-Sesma A, Simon V, Chen BK. Enhanced FCGR2A and FCGR3A signaling by HIV viremic controller IgG. JCI Insight 2017; 2:e88226. [PMID: 28239647 DOI: 10.1172/jci.insight.88226] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
HIV-1 viremic controllers (VC) spontaneously control infection without antiretroviral treatment. Several studies indicate that IgG Abs from VCs induce enhanced responses from immune effector cells. Since signaling through Fc-γ receptors (FCGRs) modulate these Ab-driven responses, here we examine if enhanced FCGR activation is a common feature of IgG from VCs. Using an infected cell-based system, we observed that VC IgG stimulated greater FCGR2A and FCGR3A activation as compared with noncontrollers, independent of the magnitude of HIV-specific Ab binding or virus neutralization activities. Multivariate regression analysis showed that enhanced FCGR signaling was a significant predictor of VC status as compared with chronically infected patients (CIP) on highly active antiretroviral therapy (HAART). Unsupervised hierarchical clustering of patient IgG functions primarily grouped VC IgG profiles by enhanced FCGR2A, FCGR3A, or dual signaling activity. Our findings demonstrate that enhanced FCGR signaling is a common and significant predictive feature of VC IgG, with VCs displaying a distinct spectrum of FCGR activation profiles. Thus, profiling FCGR activation may provide a useful method for screening and distinguishing protective anti-HIV IgG responses in HIV-infected patients and in monitoring HIV vaccination regimens.
Collapse
Affiliation(s)
- Raymond A Alvarez
- Division of Infectious Diseases, Department of Medicine, Immunology Institute
| | - Ana M Maestre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kenneth Law
- Division of Infectious Diseases, Department of Medicine, Immunology Institute
| | - Natasha D Durham
- Division of Infectious Diseases, Department of Medicine, Immunology Institute
| | - Maria Ines Barria
- Division of Infectious Diseases, Department of Medicine, Immunology Institute.,Department of Microbiology, University of Concepcion, Concepcion, Chile
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Tokyo, Japan
| | - Minoru Tada
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Tokyo, Japan
| | - Manav Kapoor
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mathew T Hotta
- Division of Infectious Diseases, Department of Medicine, Immunology Institute
| | | | - Daniel S Fierer
- Division of Infectious Diseases, Department of Medicine, Immunology Institute
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Viviana Simon
- Division of Infectious Diseases, Department of Medicine, Immunology Institute.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benjamin K Chen
- Division of Infectious Diseases, Department of Medicine, Immunology Institute
| |
Collapse
|
28
|
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
|
29
|
Choi E, Michalski CJ, Choo SH, Kim GN, Banasikowska E, Lee S, Wu K, An HY, Mills A, Schneider S, Bredeek UF, Coulston DR, Ding S, Finzi A, Tian M, Klein K, Arts EJ, Mann JFS, Gao Y, Kang CY. First Phase I human clinical trial of a killed whole-HIV-1 vaccine: demonstration of its safety and enhancement of anti-HIV antibody responses. Retrovirology 2016; 13:82. [PMID: 27894306 PMCID: PMC5126836 DOI: 10.1186/s12977-016-0317-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/17/2016] [Indexed: 01/20/2023] Open
Abstract
Background Vaccination with inactivated (killed) whole-virus particles has been used to prevent a wide range of viral diseases. However, for an HIV vaccine this approach has been largely negated due to inherent safety concerns, despite the ability of killed whole-virus vaccines to generate a strong, predominantly antibody-mediated immune response in vivo. HIV-1 Clade B NL4-3 was genetically modified by deleting the nef and vpu genes and substituting the coding sequence for the Env signal peptide with that of honeybee melittin signal peptide to produce a less virulent and more replication efficient virus. This genetically modified virus (gmHIV-1NL4-3) was inactivated and formulated as a killed whole-HIV vaccine, and then used for a Phase I human clinical trial (Trial Registration: Clinical Trials NCT01546818). The gmHIV-1NL4-3 was propagated in the A3.01 human T cell line followed by virus purification and inactivation with aldrithiol-2 and γ-irradiation. Thirty-three HIV-1 positive volunteers receiving cART were recruited for this observer-blinded, placebo-controlled Phase I human clinical trial to assess the safety and immunogenicity. Results Genetically modified and killed whole-HIV-1 vaccine, SAV001, was well tolerated with no serious adverse events. HIV-1NL4-3-specific PCR showed neither evidence of vaccine virus replication in the vaccine virus-infected human T lymphocytes in vitro nor in the participating volunteers receiving SAV001 vaccine. Furthermore, SAV001 with adjuvant significantly increased the pre-existing antibody response to HIV-1 proteins. Antibodies in the plasma of vaccinees were also found to recognize HIV-1 envelope protein on the surface of infected cells as well as showing an enhancement of broadly neutralizing antibodies inhibiting tier I and II of HIV-1 B, D, and A subtypes. Conclusion The killed whole-HIV vaccine, SAV001, is safe and triggers anti-HIV immune responses. It remains to be determined through an appropriate trial whether this immune response prevents HIV infection.
Collapse
Affiliation(s)
- Eunsil Choi
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada.,Sumagen Canada Inc., London, ON, Canada
| | - Chad J Michalski
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | | | - Gyoung Nyoun Kim
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - Elizabeth Banasikowska
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | | | - Kunyu Wu
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - Hwa-Yong An
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - Anthony Mills
- Anthony M. Mills Internal Medicine, West Hollywood, CA, USA
| | - Stefan Schneider
- Long Beach Education and Research Consultants, Long Beach, CA, USA
| | | | | | - Shilei Ding
- Centre de Recherche du CHUM, Department of Microbiology, Université de Montréal, Montreal, QC, Canada
| | - Andrés Finzi
- Centre de Recherche du CHUM, Department of Microbiology, Université de Montréal, Montreal, QC, Canada
| | - Meijuan Tian
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - Katja Klein
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - Eric J Arts
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - Jamie F S Mann
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - Yong Gao
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada
| | - C Yong Kang
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1400 Western Road, London, ON, N6G 2V4, Canada.
| |
Collapse
|
30
|
Enhanced binding of antibodies generated during chronic HIV infection to mucus component MUC16. Mucosal Immunol 2016; 9:1549-1558. [PMID: 26960182 PMCID: PMC5017893 DOI: 10.1038/mi.2016.8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/09/2016] [Indexed: 02/04/2023]
Abstract
Transmission of HIV across mucosal barriers accounts for the majority of HIV infections worldwide. Thus, efforts aimed at enhancing protective immunity at these sites are a top priority, including increasing virus-specific antibodies (Abs) and antiviral activity at mucosal sites. Mucin proteins, including the largest cell-associated mucin, mucin 16 (MUC16), help form mucus to provide a physical barrier to incoming pathogens. Here, we describe a natural interaction between Abs and MUC16 that is enhanced in specific disease settings such as chronic HIV infection. Binding to MUC16 was independent of IgG subclass, but strongly associated with shorter Ab glycan profiles, with agalactosylated (G0) Abs demonstrating the highest binding to MUC16. Binding of Abs to epithelial cells was diminished following MUC16 knockdown, and the MUC16 N-linked glycans were critical for binding. Further, agalactosylated VRC01 captured HIV more efficiently in MUC16. These data point to a novel opportunity to enrich Abs at mucosal sites by targeting Abs to MUC16 through changes in Fc glycosylation, potentially blocking viral movement and sequestering the virus far from the epithelial border. Thus, next-generation vaccines or monoclonal therapeutics may enhance protective immunity by tuning Ab glycosylation to promote the enrichment of Abs at mucosal barriers.
Collapse
|
31
|
Williams KL, Cortez V, Dingens AS, Gach JS, Rainwater S, Weis JF, Chen X, Spearman P, Forthal DN, Overbaugh J. HIV-specific CD4-induced Antibodies Mediate Broad and Potent Antibody-dependent Cellular Cytotoxicity Activity and Are Commonly Detected in Plasma From HIV-infected humans. EBioMedicine 2016; 2:1464-77. [PMID: 26629541 PMCID: PMC4634620 DOI: 10.1016/j.ebiom.2015.09.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 01/12/2023] Open
Abstract
HIV-specific antibodies (Abs) can reduce viral burden by blocking new rounds of infection or by destroying infected cells via activation of effector cells through Fc–FcR interaction. This latter process, referred to as antibody-dependent cellular cytotoxicity (ADCC), has been associated with viral control and improved clinical outcome following both HIV and SIV infections. Here we describe an HIV viral-like particle (VLP)-based sorting strategy that led to identification of HIV-specificmemory B cells encoding Abs that mediate ADCC froma subtype A-infected Kenyan woman at 914 days post-infection. Using this strategy, 12 HIV-envelope-specific monoclonal antibodies (mAbs) were isolated and three mediated potent ADCC activitywhen compared to well-characterized ADCC mAbs. The ADCC-mediating Abs also mediated antibody-dependent cell-mediated virus inhibition (ADCVI), which provides a net measure of Fc receptor-triggered effects against replicating virus. Two of the three ADCC-mediating Abs targeted a CD4-induced (CD4i) epitope also bound by the mAb C11; the third antibody targeted the N-terminus of V3. Both CD4i Abs identified here demonstrated strong cross-clade breadth with activity against 10 of 11 envelopes tested, including those from clades A, B, C, A/D and C/D, whereas the V3-specific antibody showed more limited breadth. Variants of these CD4i, C11-like mAbs engineered to interrupt binding to FcγRs inhibited a measurable percentage of the donor's ADCC activity starting as early as 189 days post-infection. C11-like antibodies also accounted for between 18–78% of ADCC activity in 9 chronically infected individuals from the same cohort study. Further, the two CD4i Abs originated from unique B cells, suggesting that antibodies targeting this epitope can be commonly produced. Taken together, these data provide strong evidence that CD4i, C11-like antibodies develop within the first 6 months of infection and they can arise fromunique B-cell lineages in the same individual. Further, thesemAbsmediate potent plasma IgG-specificADCC breadth and potency and contribute to ADCC activity in other HIV-infected individuals.
Collapse
|
32
|
Boesch AW, Brown EP, Ackerman ME. The role of Fc receptors in HIV prevention and therapy. Immunol Rev 2016; 268:296-310. [PMID: 26497529 DOI: 10.1111/imr.12339] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Over the past decade, a wealth of experimental evidence has accumulated supporting the importance of Fc receptor (FcR) ligation in antibody-mediated pathology and protection in many disease states. Here we present the diverse evidence base that has accumulated as to the importance of antibody effector functions in the setting of HIV prevention and therapy, including clinical correlates, genetic associations, viral evasion strategies, and a rapidly growing number of compelling animal model experiments. Collectively, this work identifies antibody interactions with FcR as important to both therapeutic and prophylactic strategies involving both passive and active immunity. These findings mirror those in other fields as investigators continue to work toward identifying the right antibodies and the right effectors to be present at the right sites at the right time.
Collapse
Affiliation(s)
- Austin W Boesch
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.,Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH, USA.,Department of Microbiology and Immunology, Geisel School of Medicine, Lebanon, NH, USA
| |
Collapse
|
33
|
de Souza MS, Pinyakorn S, Akapirat S, Pattanachaiwit S, Fletcher JLK, Chomchey N, Kroon ED, Ubolyam S, Michael NL, Robb ML, Phanuphak P, Kim JH, Phanuphak N, Ananworanich J. Initiation of Antiretroviral Therapy During Acute HIV-1 Infection Leads to a High Rate of Nonreactive HIV Serology. Clin Infect Dis 2016; 63:555-61. [PMID: 27317797 DOI: 10.1093/cid/ciw365] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/23/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Third- and fourth-generation immunoassays (IAs) are widely used in the diagnosis of human immunodeficiency virus (HIV) infection. Antiretroviral therapy (ART) during acute HIV infection (AHI) may impact HIV-specific antibodies, with failure to develop antibody or seroreversion. We report on the ability of diagnostic tests to detect HIV-specific antibodies in Thai participants initiating ART during AHI. METHODS Participants with detectable plasma HIV RNA but nonreactive HIV-specific immunoglobulin G, enrolled in an AHI study, were offered immediate initiation of ART. Participants were tested at initiation and at 12 and 24 weeks following treatment using standard second-, third-, and fourth-generation IAs and Western blot (WB). RESULTS Participants (N = 234) initiating ART at a median of 19 days (range, 1-62 days) from HIV exposure demonstrated different frequencies of reactivity prior to and following 24 weeks of ART depending on the IA. Third-generation IA nonreactivity prior to ART was 48%, which decreased to 4% following ART (P < .001). Fourth-generation IA nonreactivity was 18% prior to ART and 17% following ART (P = .720). Negative WB results were observed in 89% and 12% of participants prior to and following 24 weeks of ART, respectively (P < .001). Seroreversion to nonreactivity during ART was observed to at least one of the tests in 20% of participants, with fourth-generation IA demonstrating the highest frequency (11%) of seroreversion. CONCLUSIONS HIV-specific antibodies may fail to develop and, when detected, may decline when ART is initiated during AHI. Although fourth-generation IA was the most sensitive at detecting AHI prior to ART, third-generation IA was the most sensitive during treatment. CLINICAL TRIALS REGISTRATION NCT00796146 and NCT00796263.
Collapse
Affiliation(s)
- Mark S de Souza
- South East Asia Research Collaboration with Hawaii (SEARCH) Thai Red Cross AIDS Research Centre, Bangkok, Thailand Cooper Human Systems, Cambridge, Massachusetts
| | - Suteeraporn Pinyakorn
- Henry M. Jackson Foundation for the Advancement of Military Medicine United States Military HIV Research Program, Bethesda, Maryland
| | - Siriwat Akapirat
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences, United States Component
| | | | | | | | - Eugene D Kroon
- South East Asia Research Collaboration with Hawaii (SEARCH) Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | | | - Nelson L Michael
- United States Military HIV Research Program, Bethesda, Maryland Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Merlin L Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine United States Military HIV Research Program, Bethesda, Maryland
| | - Praphan Phanuphak
- South East Asia Research Collaboration with Hawaii (SEARCH) Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Jerome H Kim
- International Vaccine Institute, Seoul, South Korea
| | - Nittaya Phanuphak
- South East Asia Research Collaboration with Hawaii (SEARCH) Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Jintanat Ananworanich
- South East Asia Research Collaboration with Hawaii (SEARCH) Henry M. Jackson Foundation for the Advancement of Military Medicine United States Military HIV Research Program, Bethesda, Maryland
| | | |
Collapse
|
34
|
Comparison of Antibody-Dependent Cell-Mediated Cytotoxicity and Virus Neutralization by HIV-1 Env-Specific Monoclonal Antibodies. J Virol 2016; 90:6127-6139. [PMID: 27122574 DOI: 10.1128/jvi.00347-16] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/16/2016] [Indexed: 01/07/2023] Open
Abstract
UNLABELLED Although antibodies to the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein have been studied extensively for their ability to block viral infectivity, little data are currently available on nonneutralizing functions of these antibodies, such as their ability to eliminate virus-infected cells by antibody-dependent cell-mediated cytotoxicity (ADCC). HIV-1 Env-specific antibodies of diverse specificities, including potent broadly neutralizing and nonneutralizing antibodies, were therefore tested for ADCC against cells infected with a lab-adapted HIV-1 isolate (HIV-1NL4-3), a primary HIV-1 isolate (HIV-1JR-FL), and a simian-human immunodeficiency virus (SHIV) adapted for pathogenic infection of rhesus macaques (SHIVAD8-EO). In accordance with the sensitivity of these viruses to neutralization, HIV-1NL4-3-infected cells were considerably more sensitive to ADCC, both in terms of the number of antibodies and magnitude of responses, than cells infected with HIV-1JR-FL or SHIVAD8-EO ADCC activity generally correlated with antibody binding to Env on the surfaces of virus-infected cells and with viral neutralization; however, neutralization was not always predictive of ADCC, as instances of ADCC in the absence of detectable neutralization, and vice versa, were observed. These results reveal incomplete overlap in the specificities of antibodies that mediate these antiviral activities and provide insights into the relationship between ADCC and neutralization important for the development of antibody-based vaccines and therapies for combating HIV-1 infection. IMPORTANCE This study provides fundamental insights into the relationship between antibody-dependent cell-mediated cytotoxicity (ADCC) and virus neutralization that may help to guide the development of antibody-based vaccines and immunotherapies for the prevention and treatment of HIV-1 infection.
Collapse
|
35
|
Wikramaratna PS, Lourenço J, Klenerman P, Pybus OG, Gupta S. Effects of neutralizing antibodies on escape from CD8+ T-cell responses in HIV-1 infection. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0290. [PMID: 26150656 PMCID: PMC4528488 DOI: 10.1098/rstb.2014.0290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite substantial advances in our knowledge of immune responses against HIV-1 and of its evolution within the host, it remains unclear why control of the virus eventually breaks down. Here, we present a new theoretical framework for the infection dynamics of HIV-1 that combines antibody and CD8+ T-cell responses, notably taking into account their different lifespans. Several apparent paradoxes in HIV pathogenesis and genetics of host susceptibility can be reconciled within this framework by assigning a crucial role to antibody responses in the control of viraemia. We argue that, although escape from or progressive loss of quality of CD8+ T-cell responses can accelerate disease progression, the underlying cause of the breakdown of virus control is the loss of antibody induction due to depletion of CD4+ T cells. Furthermore, strong antibody responses can prevent CD8+ T-cell escape from occurring for an extended period, even in the presence of highly efficacious CD8+ T-cell responses.
Collapse
Affiliation(s)
| | - José Lourenço
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK
| | - Paul Klenerman
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK
| | - Sunetra Gupta
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK
| |
Collapse
|
36
|
He X, Simoneau CR, Granoff ME, Lunemann S, Dugast AS, Shao Y, Altfeld M, Körner C. Assessment of the antiviral capacity of primary natural killer cells by optimized in vitro quantification of HIV-1 replication. J Immunol Methods 2016; 434:53-60. [PMID: 27094484 DOI: 10.1016/j.jim.2016.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 10/21/2022]
Abstract
Despite a growing number of studies investigating the impact of natural killer (NK) cells on HIV-1 pathogenesis, the exact mechanism by which NK cells recognize HIV-1-infected cells and exert immunological pressure on HIV-1 remains unknown. Previously several groups including ours have introduced autologous HIV-1-infected CD4(+) T cells as suitable target cells to study NK-cell function in response to HIV-1 infection in vitro. Here, we re-evaluated and optimized a standardized in vitro assay that allows assessing the antiviral capacity of NK cells. This includes the implementation of HIV-1 RNA copy numbers as readout for NK-cell-mediated inhibition of HIV-1 replication and the investigation of inter-assay variation in comparison to previous methods, such as HIV-1 p24 Gag production and frequency of p24(+) CD4(+) T cells. Furthermore, we investigated the possibility to hasten the duration of the assay and provide concepts for downstream applications. Autologous CD4(+) T cells and NK cells were obtained from peripheral blood of HIV-negative healthy individuals and were separately enriched through negative selection. CD4(+) T cells were infected with the HIV-1 strain JR-CSF at an MOI of 0.01. Infected CD4(+) T cells were then co-cultured with primary NK cells at various effector:target ratios for up to 14days. Supernatants obtained from media exchanged at days 4, 7, 11 and 14 were used for quantification of HIV-1 p24 Gag and HIV-1 RNA copy numbers. In addition, frequency of infected CD4(+) T cells was determined by flow cytometric detection of intracellular p24 Gag. The assay displayed minimal inter-assay variation when utilizing viral RNA quantification or p24 Gag concentration for the assessment of viral replication. Viral RNA quantification was more rigorous to display magnitude and kinetics of NK-cell-mediated inhibition of HIV-1 replication, longitudinally and between tested individuals. The results of this study demonstrate that NK-cell-mediated inhibition of HIV-1 replication can be reliably quantified in vitro, and that viral RNA quantification is comparable to p24 Gag quantification via ELISA, providing a robust measurement for NK-cell-mediated inhibition of viral replication. Overall, the described assay provides an optimized tool to study the antiviral capacity of NK cells against HIV-1 and an additional experimental tool to investigate the molecular determinants of NK-cell recognition of virus-infected cells.
Collapse
Affiliation(s)
- Xuan He
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA; State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, People's Republic of China.
| | - Camille R Simoneau
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA.
| | - Mitchell E Granoff
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA.
| | - Sebastian Lunemann
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251 Hamburg, Germany.
| | - Anne-Sophie Dugast
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA.
| | - Yiming Shao
- State Key Laboratory for Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Changping District, Beijing 102206, People's Republic of China.
| | - Marcus Altfeld
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA; Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251 Hamburg, Germany.
| | - Christian Körner
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA; Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251 Hamburg, Germany.
| |
Collapse
|
37
|
Env-Specific IgA from Viremic HIV-Infected Subjects Compromises Antibody-Dependent Cellular Cytotoxicity. J Virol 2016; 90:670-81. [PMID: 26491172 PMCID: PMC4702681 DOI: 10.1128/jvi.02363-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/14/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Elucidating the factors that modulate HIV-specific antibody-dependent cellular cytotoxicity (ADCC) will help in understanding its role in HIV immunity. The aim of this study was to determine whether IgA could modify the magnitude of ADCC in HIV infection, abrogating its protective role. Plasma samples from 20 HIV-positive (HIV(+)) subjects enrolled during primary HIV infection (PHI), 10 chronically infected subjects (chronic), and 7 elite controllers (EC) were used. ADCC was determined by using a fluorometric ADCC assay, before and after removal of plasma IgA. Data were analyzed by using nonparametric statistics. ADCC was documented in 80% of PHI enrollment samples and in 100% of PHI 12-month, chronic, and EC samples; it peaked after acute infection, reached a plateau in chronic infection, and decreased after initiation of antiretroviral treatment (ART). Significant associations between ADCC and disease progression were found only after removal of plasma IgA from 12-month PHI samples: the magnitude of ADCC not only increased after IgA removal but also correlated with CD4(+) T-cell preservation. This work provides evidence that gp120-specific IgA was capable of modifying ADCC responses during natural HIV infection for the first time and adds to similar evidence provided in other settings. Furthermore, it underscores the complexity of the ADCC phenomenon and will help in an understanding of its underlying mechanisms. IMPORTANCE Although the induction of ADCC-mediating antibodies in HIV-infected subjects has been extensively documented, the association of these antibodies with protection from disease progression is poorly understood. Here, we demonstrate that plasma IgA is a factor capable of modifying the magnitude of IgG-mediated ADCC in HIV infection, mitigating its beneficial effect. These results help in understanding why previous studies failed to demonstrate correlations between ADCC and disease progression, and they also contribute to the notion that an HIV vaccine should stimulate the production of ADCC-mediating IgG antibodies but not IgA.
Collapse
|
38
|
Ackerman ME, Mikhailova A, Brown EP, Dowell KG, Walker BD, Bailey-Kellogg C, Suscovich TJ, Alter G. Polyfunctional HIV-Specific Antibody Responses Are Associated with Spontaneous HIV Control. PLoS Pathog 2016; 12:e1005315. [PMID: 26745376 PMCID: PMC4706315 DOI: 10.1371/journal.ppat.1005315] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/09/2015] [Indexed: 12/31/2022] Open
Abstract
Elite controllers (ECs) represent a unique model of a functional cure for HIV-1 infection as these individuals develop HIV-specific immunity able to persistently suppress viremia. Because accumulating evidence suggests that HIV controllers generate antibodies with enhanced capacity to drive antibody-dependent cellular cytotoxicity (ADCC) that may contribute to viral containment, we profiled an array of extra-neutralizing antibody effector functions across HIV-infected populations with varying degrees of viral control to define the characteristics of antibodies associated with spontaneous control. While neither the overall magnitude of antibody titer nor individual effector functions were increased in ECs, a more functionally coordinated innate immune-recruiting response was observed. Specifically, ECs demonstrated polyfunctional humoral immune responses able to coordinately recruit ADCC, other NK functions, monocyte and neutrophil phagocytosis, and complement. This functionally coordinated response was associated with qualitatively superior IgG3/IgG1 responses, whereas HIV-specific IgG2/IgG4 responses, prevalent among viremic subjects, were associated with poorer overall antibody activity. Rather than linking viral control to any single activity, this study highlights the critical nature of functionally coordinated antibodies in HIV control and associates this polyfunctionality with preferential induction of potent antibody subclasses, supporting coordinated antibody activity as a goal in strategies directed at an HIV-1 functional cure.
Collapse
Affiliation(s)
- Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
- * E-mail: (MEA); (GA)
| | - Anastassia Mikhailova
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Eric P. Brown
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Karen G. Dowell
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Chris Bailey-Kellogg
- Department of Computer Science, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Todd J. Suscovich
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
- * E-mail: (MEA); (GA)
| |
Collapse
|
39
|
Prospects for engineering HIV-specific antibodies for enhanced effector function and half-life. Curr Opin HIV AIDS 2016; 10:160-9. [PMID: 25700208 DOI: 10.1097/coh.0000000000000149] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE OF REVIEW A wealth of recent animal model data suggests that as exciting possibilities for the use of antibodies in passive immunotherapy strategies continue to develop, it will be important to broadly consider how antibodies achieve anti-HIV-1 effect in vivo. RECENT FINDINGS Beyond neutralization breadth and potency, substantial evidence from natural infection, vaccination, and studies in animal models points to a critical role for antibody Fc receptor (FcR) engagement in reducing risk of infection, decreasing postinfection viremia, and delaying viral rebound. Supporting these findings in the setting of HIV, the clinical maturation of recombinant antibody therapeutics has reinforced the importance of Fc-driven activity in vivo across many disease settings, as well as opportunely resulted in the development and exploration of a number of engineered Fc sequence and glycosylation variants that possess differential binding to FcRs. Exploiting these variants as tools, the individual and concerted effects of antibody effector functions such as antibody-dependent cellular cytotoxicity, antibody-dependent cell-mediated virus inhibition, phagocytosis, complement-dependent cytotoxicity, antibody half-life, and compartmentalization are now being explored. As exciting molecular therapies are advanced, these studies promise to provide insight into optimal in-vivo antibody activity profiles. SUMMARY Careful consideration of recent progress in understanding protective antibody activities in vivo can point toward how tailoring antibody activity via Fc domain modification may enable optimization of HIV prevention and eradication strategies.
Collapse
|
40
|
Milligan C, Richardson BA, John-Stewart G, Nduati R, Overbaugh J. Passively acquired antibody-dependent cellular cytotoxicity (ADCC) activity in HIV-infected infants is associated with reduced mortality. Cell Host Microbe 2016; 17:500-6. [PMID: 25856755 DOI: 10.1016/j.chom.2015.03.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/11/2014] [Accepted: 02/18/2015] [Indexed: 12/11/2022]
Abstract
In addition to direct effects on virus infectivity, antibodies mediate antibody-dependent cellular cytotoxicity (ADCC), the killing of an antibody-coated virus-infected cell by cytotoxic effector cells. Although ADCC has been suggested to protect against HIV, the relationship between HIV-specific ADCC antibodies at the time of HIV exposure and infection outcome in humans remains to be assessed. We evaluated the ADCC activity of passively acquired antibodies in infants born to HIV-infected mothers. ADCC levels were higher in uninfected than infected infants, although not significantly. Increase in ADCC antibody activity in infected infants was associated with reduced mortality risk. Infant ADCC positively correlated with the magnitude of IgG1 binding, and IgG1 levels were associated with survival in infected infants. Infant IgG3-binding antibodies were not associated with infected infant survival. These data suggest a therapeutic benefit of pre-existing HIV-specific ADCC antibodies and support a role for eliciting ADCC-mediating IgG1 in HIV vaccines.
Collapse
Affiliation(s)
- Caitlin Milligan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Barbra A Richardson
- Department of Global Health, University of Washington, Seattle, WA 98195, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Ruth Nduati
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Julie Overbaugh
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Contrasting Adult and Infant Immune Responses to HIV Infection and Vaccination. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 23:84-94. [PMID: 26656117 DOI: 10.1128/cvi.00565-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Extensive studies have demonstrated that infant immune responses are distinct from those of adults. Despite these differences, infant immunization can elicit protective immune responses at levels comparable to or, in some cases, higher than adult immune responses to many vaccines. To date, only a few HIV vaccine candidates have been tested in infant populations, and none of them evaluated vaccine efficacy. Recent exciting studies showing that HIV-infected infants can develop broad neutralizing antibody responses and that some HIV vaccine regimens can elicit high levels of potentially protective antibodies in infants provide support for the development and testing of HIV vaccines in pediatric populations. In this review, we discuss the differences in adult and infant immune responses in the setting of HIV infection and vaccination.
Collapse
|
42
|
Sadanand S, Suscovich TJ, Alter G. Broadly Neutralizing Antibodies Against HIV: New Insights to Inform Vaccine Design. Annu Rev Med 2015; 67:185-200. [PMID: 26565674 DOI: 10.1146/annurev-med-091014-090749] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
HIV-1 poses immense immunological challenges to the humoral immune response because of its ability to shield itself and replicate and evolve rapidly. Although most currently licensed vaccines provide protection via the induction of antibodies (Abs) that can directly block infection ( 1 ), 30 years of HIV-1 vaccine research has failed to successfully elicit such Abs against globally relevant HIV strains. However, mounting evidence suggests that these broadly neutralizing antibodies (bNAbs) do emerge naturally in a significant fraction of infected subjects, albeit after years of infection, indicating that these responses can be selected naturally by the immune response but take long periods of time to evolve. We review the basic structural characteristics of broadly neutralizing antibodies and how they recognize the virus, and we discuss new vaccination strategies that aim to mimic natural evolution to guide B cells to produce protective Abs against HIV-1.
Collapse
Affiliation(s)
- Saheli Sadanand
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139-3583; , ,
| | - Todd J Suscovich
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139-3583; , ,
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139-3583; , ,
| |
Collapse
|
43
|
Alexander MR, Sanders RW, Moore JP, Klasse PJ. Short Communication: Virion Aggregation by Neutralizing and Nonneutralizing Antibodies to the HIV-1 Envelope Glycoprotein. AIDS Res Hum Retroviruses 2015; 31:1160-5. [PMID: 26086186 DOI: 10.1089/aid.2015.0050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We used dynamic light scattering to detect aggregation of HIV-1 virions by antibodies (IgG) to the viral envelope glycoprotein (Env). Virions of different strains were inactivated by 2,2'-dithiodipyridine (AT-2), a procedure that abrogates infectivity but preserves the native antigenic structure of Env. Neutralizing antibodies directed to a V3-base- and glycan-dependent epitope on gp120 and to the apex of the Env trimer, as well as nonneutralizing antibodies to the epitope cluster I on the gp41-ectodomain, aggregated virions, but in markedly narrow concentration ranges. In contrast, the neutralizing antibody 2G12, which is specific for a composite glycan-dependent epitope on gp120 and functionally monovalent because of its unusual domain-swap structure, was nonaggregating. These results have potentially complex implications for vaccine development.
Collapse
Affiliation(s)
- Marina R. Alexander
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| | - Rogier W. Sanders
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| | - Per Johan Klasse
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| |
Collapse
|
44
|
Nimmerjahn F. A constant threat for HIV: Fc-engineering to enhance broadly neutralizing antibody activity for immunotherapy of the acquired immunodeficiency syndrome. Eur J Immunol 2015; 45:2183-90. [PMID: 26140474 DOI: 10.1002/eji.201445386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 01/12/2023]
Abstract
Passive immunotherapy with polyclonal or hyperimmune serum immunoglobulin G (IgG) preparations provides an efficient means of protecting immunocompromised patients from microbial infections. More recently, the use of passive immunotherapy to prevent or to treat established infections with the human immunodeficiency virus (HIV) has gained much attention, due to promising preclinical data obtained in monkey and humanized mouse in vivo model systems, demonstrating that the transfer of HIV-specific antibodies can not only prevent HIV infection, but also diminish virus load during chronic infection. Furthermore, an array of broadly neutralizing HIV-specific antibodies has become available and the importance of the IgG constant region as a critical modulator of broadly neutralizing activity has been demonstrated. The aim of this review is to summarize the most recent findings with regard to the molecular and cellular mechanisms responsible for antibody-mediated clearance of HIV infection, and to discuss how this may help to improve HIV therapy via optimizing Fcγ-receptor-dependent activities of HIV-specific antibodies.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Institute of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
45
|
Lee WS, Parsons MS, Kent SJ, Lichtfuss M. Can HIV-1-Specific ADCC Assist the Clearance of Reactivated Latently Infected Cells? Front Immunol 2015; 6:265. [PMID: 26074924 PMCID: PMC4445400 DOI: 10.3389/fimmu.2015.00265] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/13/2015] [Indexed: 02/05/2023] Open
Affiliation(s)
- Wen Shi Lee
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, VIC , Australia
| | - Matthew Sidney Parsons
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, VIC , Australia
| | - Stephen John Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, VIC , Australia
| | - Marit Lichtfuss
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne, VIC , Australia
| |
Collapse
|