25
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Vaccine-Induced, High-Magnitude HIV Env-Specific Antibodies with Fc-Mediated Effector Functions Are Insufficient to Protect Infant Rhesus Macaques against Oral SHIV Infection

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          ABSTRACT

          Improved access to antiretroviral therapy (ART) and antenatal care has significantly reduced in utero and peripartum mother-to-child human immunodeficiency virus (HIV) transmission. However, as breast milk transmission of HIV still occurs at an unacceptable rate, there remains a need to develop an effective vaccine for the pediatric population. Previously, we compared different HIV vaccine strategies, intervals, and adjuvants in infant rhesus macaques to optimize the induction of HIV envelope (Env)-specific antibodies with Fc-mediated effector function. In this study, we tested the efficacy of an optimized vaccine regimen against oral simian-human immunodeficiency virus (SHIV) acquisition in infant macaques. Twelve animals were immunized with 1086.c gp120 protein adjuvanted with 3M-052 in stable emulsion and modified vaccinia Ankara (MVA) virus expressing 1086.c HIV Env. Twelve control animals were immunized with empty MVA. The vaccine prime was given within 10 days of birth, with booster doses being administered at weeks 6 and 12. The vaccine regimen induced Env-specific plasma IgG antibodies capable of antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). Beginning at week 15, infants were exposed orally to escalating doses of heterologous SHIV-1157(QNE)Y173H once a week until infected. Despite the induction of strong Fc-mediated antibody responses, the vaccine regimen did not reduce the risk of infection or time to acquisition compared to controls. However, among vaccinated animals, ADCC postvaccination and postinfection was associated with reduced peak viremia. Thus, nonneutralizing Env-specific antibodies with Fc effector function elicited by this vaccine regimen were insufficient for protection against heterologous oral SHIV infection shortly after the final immunization but may have contributed to control of viremia.

          IMPORTANCE Women of childbearing age are three times more likely to contract HIV infection than their male counterparts. Poor HIV testing rates coupled with low adherence to antiretroviral therapy (ART) result in a high risk of mother-to-infant HIV transmission, especially during the breastfeeding period. A preventative vaccine could curb pediatric HIV infections, reduce potential health sequalae, and prevent the need for lifelong ART in this population. The results of the current study imply that the HIV Env-specific IgG antibodies elicited by this candidate vaccine regimen, despite a high magnitude of Fc-mediated effector function but a lack of neutralizing antibodies and polyfunctional T cell responses, were insufficient to protect infant rhesus macaques against oral virus acquisition.

          Related collections

          Most cited references88

          • Record: found
          • Abstract: found
          • Article: not found

          Innate immunity of the newborn: basic mechanisms and clinical correlates.

          Ofer Levy (2007)
          The fetus and newborn face a complex set of immunological demands, including protection against infection, avoidance of harmful inflammatory immune responses that can lead to pre-term delivery, and balancing the transition from a sterile intra-uterine environment to a world that is rich in foreign antigens. These demands shape a distinct neonatal innate immune system that is biased against the production of pro-inflammatory cytokines. This bias renders newborns at risk of infection and impairs responses to many vaccines. This Review describes innate immunity in newborns and discusses how this knowledge might be used to prevent and treat infection in this vulnerable population.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Immune-correlates analysis of an HIV-1 vaccine efficacy trial.

            In the RV144 trial, the estimated efficacy of a vaccine regimen against human immunodeficiency virus type 1 (HIV-1) was 31.2%. We performed a case-control analysis to identify antibody and cellular immune correlates of infection risk. In pilot studies conducted with RV144 blood samples, 17 antibody or cellular assays met prespecified criteria, of which 6 were chosen for primary analysis to determine the roles of T-cell, IgG antibody, and IgA antibody responses in the modulation of infection risk. Assays were performed on samples from 41 vaccinees who became infected and 205 uninfected vaccinees, obtained 2 weeks after final immunization, to evaluate whether immune-response variables predicted HIV-1 infection through 42 months of follow-up. Of six primary variables, two correlated significantly with infection risk: the binding of IgG antibodies to variable regions 1 and 2 (V1V2) of HIV-1 envelope proteins (Env) correlated inversely with the rate of HIV-1 infection (estimated odds ratio, 0.57 per 1-SD increase; P=0.02; q=0.08), and the binding of plasma IgA antibodies to Env correlated directly with the rate of infection (estimated odds ratio, 1.54 per 1-SD increase; P=0.03; q=0.08). Neither low levels of V1V2 antibodies nor high levels of Env-specific IgA antibodies were associated with higher rates of infection than were found in the placebo group. Secondary analyses suggested that Env-specific IgA antibodies may mitigate the effects of potentially protective antibodies. This immune-correlates study generated the hypotheses that V1V2 antibodies may have contributed to protection against HIV-1 infection, whereas high levels of Env-specific IgA antibodies may have mitigated the effects of protective antibodies. Vaccines that are designed to induce higher levels of V1V2 antibodies and lower levels of Env-specific IgA antibodies than are induced by the RV144 vaccine may have improved efficacy against HIV-1 infection.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Beyond binding: antibody effector functions in infectious diseases

              Antibodies play an essential role in host defence against pathogens by recognizing microorganisms or infected cells. Although preventing pathogen entry is one potential mechanism of protection, antibodies can control and eradicate infections through a variety of other mechanisms. In addition to binding and directly neutralizing pathogens, antibodies drive the clearance of bacteria, viruses, fungi and parasites via their interaction with the innate and adaptive immune systems, leveraging a remarkable diversity of antimicrobial processes locked within our immune system. Specifically, antibodies collaboratively form immune complexes that drive sequestration and uptake of pathogens, clear toxins, eliminate infected cells, increase antigen presentation and regulate inflammation. The diverse effector functions that are deployed by antibodies are dynamically regulated via differential modification of the antibody constant domain, which provides specific instructions to the immune system. Here, we review mechanisms by which antibody effector functions contribute to the balance between microbial clearance and pathology and discuss tractable lessons that may guide rational vaccine and therapeutic design to target gaps in our infectious disease armamentarium.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                mSphere
                mSphere
                msphere
                mSphere
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2379-5042
                23 February 2022
                Jan-Feb 2022
                23 February 2022
                : 7
                : 1
                : e00839-21
                Affiliations
                [a ] Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hillgrid.10698.36, , Chapel Hill, North Carolina, USA
                [b ] Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hillgrid.10698.36, , Chapel Hill, North Carolina, USA
                [c ] Children’s Research Institute, School of Medicine, University of North Carolina at Chapel Hillgrid.10698.36, , Chapel Hill, North Carolina, USA
                [d ] Department of Biostatistics, Gillings School of Public Health, University of North Carolina at Chapel Hillgrid.10698.36, , Chapel Hill, North Carolina, USA
                [e ] Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
                [f ] Department of Surgery, Duke University School of Medicinegrid.471396.e, , Durham, North Carolina, USA
                [g ] Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
                [h ] Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
                [i ] California National Primate Research Center, University of California at Davis, Davis, California, USA
                [j ] Department of Pediatrics, Weill Cornell Medical College, New York, New York, USA
                University of Florida
                Author notes

                The authors declare no conflict of interest.

                Author information
                https://orcid.org/0000-0002-2317-6311
                https://orcid.org/0000-0003-0941-0703
                https://orcid.org/0000-0002-0960-1019
                https://orcid.org/0000-0002-7375-1337
                https://orcid.org/0000-0001-5199-3004
                Article
                00839-21 msphere.00839-21
                10.1128/msphere.00839-21
                8865927
                35196125
                fdc930e6-f23f-492c-bb91-ad9e93c25f22
                Copyright © 2022 Curtis et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 9 October 2021
                : 4 February 2022
                Page count
                supplementary-material: 1, Figures: 8, Tables: 5, Equations: 0, References: 89, Pages: 21, Words: 14253
                Funding
                Funded by: National Institute of Allergy and Infectious Diseases (NIAID), FundRef https://doi.org/10.13039/100000060;
                Award ID: P01 AI117915
                Award Recipient :
                Funded by: Center for AIDS Research, University of North Carolina at Chapel HIll, FundRef https://doi.org/10.13039/100011072;
                Award ID: 5P30AI050410
                Award Recipient :
                Categories
                Research Article
                vaccines, Vaccines
                Custom metadata
                January/February 2022

                adcc,fc-mediated antibody function,pediatric hiv vaccine,rhesus macaque

                Comments

                Comment on this article