35
views
0
recommends
+1 Recommend
0 collections
    8
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Design Challenges for HIV-1 Vaccines Based on Humoral Immunity

      review-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

          Introduction Human Immunodeficiency Virus-1 (HIV-1) presents profound challenges to vaccine developers. Potential hurdles of relevance to the design of an effective HIV-1 vaccine based on humoral immunity include: (1) the exceptional rate of mutation of the viral genome due to an error-prone polymerase (i.e., reverse transcriptase) (1), (2) a relatively high rate of genomic recombination (2, 3), (3) both within-host and between-host evolution (4), (4) extensive glycosylation of the chief antigenic target (gp120) recognized by potentially protective antibodies (5, 6), (5) immunodominance of regions of the envelope glycoprotein that display a high degree of primary structure diversity (7) favoring the generation of neutralizing antibodies of narrow breadth, (6) a relatively low density of antigen spikes on the virion surface (8, 9) thereby minimizing multivalent antibody binding and perhaps raising the threshold affinity required for potent neutralization by at least some antibodies, (7) the ability of HIV to infect CD4+ T cells and other cells critical to functioning of the immune system, and deplete the numbers of these cells, and (8) perhaps most insidious, activation of CD4+ T cells, which is necessary for the generation of potent broadly neutralizing antibodies in response to immunization may simultaneously increase the number of cells susceptible to infection by HIV (10, 11). The scale of the antigenic diversity characterizing the HIV-1 envelope molecules, which are the primary targets of antibody-mediated immunity, is truly daunting. According to Korber et al. (1), the HIV-1 viral genomes in one infected individual encompass the same approximate extent of nucleotide sequence diversity that is exhibited by the worldwide population of influenza A viral genomes over the course of a year. While most HIV transmission events appear to trace back to a single virus, up to a quarter of infections may involve infection by two to five viruses (12). So, even if vaccine immunization elicits a robust antibody response, the probability that the antibodies circulating in the blood of a vaccinated individual will effectively neutralize or otherwise mediate immunity against all of the viruses mediating infection will be greatest if those antibodies are broadly neutralizing. In addition, due to rapid within-host evolution, substantially different viruses may be infecting different individuals in a large population. Therefore, antibodies generated by vaccination will need to be broadly neutralizing to achieve high levels of vaccine efficacy. Unique Features of Broadly Neutralizing Antibodies for HIV-1 Investigators interested in HIV-1 vaccines based on humoral immunity have established that most neutralizing antibodies in most patients neutralize a limited range of strains, primarily those to which the individual patient was exposed or closely related strains. In contrast, only about 25–30% of patients synthesize potent and broadly neutralizing antibodies (pbnAb) and typically only after 2–4 years following the initiation of infection. This time interval for the development of protective antibodies is exceptionally long. The extent of somatic hypermutation focused on the rearranged immunoglobulin (Ig) genes encoding the heavy and light chain variable domains and that has been associated with broad neutralizing activity against HIV-1 is also extraordinary. Ig genes encoding typical anti-pathogen antibodies from a mature immune response may exhibit on the order of 15–20 mutations in the gene segments encoding the heavy chain variable domain. In contrast, many pbnAb for HIV-1 have 40–100 somatic mutations in the genes encoding the heavy chain variable domain. It seems plausible to infer that a truly exceptional extent of Ig gene evolution is contributory to and possibly essential for the generation of at least some pbnAb. This inference is supported by the recent findings that many pbnAb contain many heavy chain V domain-encoding mutations in framework regions and these mutations, at both contact and non-contact positions, are necessary for both high potency and great breadth of neutralization activity (13). Although, the high number of variable domain mutations in pbnAb may also reflect in part the rapid pace of within-host HIV evolution, this possibility does not obviously account for the typical and rather extended time frame observed for the development of pbnAb. In either case, it will also be important to study the critical role of follicular helper CD4+ cells (14) in driving proliferation as well as somatic hypermutation and affinity maturation in germinal center B cells. Another useful focus for future investigation is based on the recent insights regarding the neutralizing activities of glycan-specific Ab (15). Thus, it is fair to question whether the standard approach to clinical vaccination of administering the same immunogen several times is likely to be effective in the case of HIV-1. A number of major laboratories investigating this problem have already begun fashioning novel approaches in which the ultimate overall goal is likely to be immunization with a series of related but distinguishable HIV-1-derived envelope molecules that guide the evolutionary trajectory for the genes encoding the variable (V) domains of the corresponding neutralizing antibodies (16). Such a scheme would appear to recapitulate to some extent the in vivo process that occurs for B cells in infected patients. The logistics of vaccine delivery entailed by this sort of immunization scheme may be more challenging than for any previously successful clinical vaccine. A crucial feature of this approach of guiding B cell evolution to the production of pbnAb reflects the troubling fact that the epitope recognized by a protective Ab at the end of the process may not be bound by the B cell receptors (BCR) of the germline B cells from which such antibodies will ultimately be derived by clonal evolution (16). Therefore, vaccine developers have begun working to identify antigens suitable for the activation of the B cells expressing germline BCRs that can serve as the ancestral sequences for pbnAb. This sort of approach is undoubtedly made more plausible by recent and impressive advances in structure-guided methods for designing vaccine immunogens to contain specific epitopes (16, 17). The results of Dey et al. and Jardine et al. suggest the potential of these new techniques for eliciting antibody responses of desired specificity for the HIV-1 envelope protein. Remaining Questions Regarding Immunity Elicited by Vaccine-Guided B Cell Evolution Nevertheless, it remains to be determined: (1) if identification of monoclonal pbnAb can lead to designed vaccine immunogens that reliably elicit protective polyclonal serum or mucosal antibodies, (2) how many sequential immunogens will be required to reliably guide the evolutionary trajectory to pbnAb in the great majority of vaccine recipients, (3) how many total immunizations will be needed, (4) what intervals between immunizations would achieve the optimal balance between immunogenicity and a timeframe for elicitation of protective responses compatible with public health goals, and (5) how much expense such a scheme, implemented on a mass scale, will entail. The preceding strategy is based on the assumption that the most viable approach to generating effective immunity to HIV-1 is to elicit pbnAb by an active humoral immune response. There is evidence that non-neutralizing antibodies may be able to contribute to immunity to the virus (18–20), but it appears that only a minority of investigators focusing on HIV vaccines based on humoral immunity are persuaded that such immunity can be of sufficient potency to protect vaccine recipients in the absence of pbnAb. It is of importance of determine if elicitation of non-neutralizing antibodies to HIV-1 antigens is a feasible path to successful protection in a high percentage of vaccine recipients. Regulating CD4+ T Cells as a Path to Immunity to HIV-1 A counter-intuitive approach to vaccine development, although not based in elicitation of humoral immunity, merits brief discussion. Since HIV-1 infects CD4+ T cells (which are crucial for the sorts of humoral immune responses we address above), and susceptibility to HIV-1 is increased after activation through the T cell receptor, Lu et al. (10) reasoned that an immunogen able to diminish responses of CD4+ T cells might actually reduce susceptibility of a recipient of that immunogen to HIV infection. These authors demonstrated that administration of an oral vaccine consisting of an inactivated simian immunodeficiency virus (SIVmac239) plus the tolerance-inducing commensal bacterium Lactobacillus plantarum to macaques protected them from subsequent intrarectal challenge. CD8+ T cell-depleting antibodies confirmed the necessary role of CD8+ regulatory T cells to this unusual form of vaccine-mediated immunity to infection. In the macaque model of SIV infection described by Lu et al., the beneficial effect of reduced responsiveness by CD4+ T cells was mediated by CD8+ T cells recognizing antigens presented by non-classical MHC class I molecules. It will obviously be of interest to determine if the same cell type could operate similarly in humans, assuming the phenomenon is reproducible across species. Another important question that merits investigation is whether this approach to vaccination is effective in eliciting protection against infection for virus entry by other routes. Even if in humans, a vaccine targeting CD8+ T cells recognizing antigens presented by non-classical MHC class I molecules were able to provide protection from HIV infection, it would not necessarily rule out the possibility of manipulating standard CD4+ regulatory T cells to add to any benefit associated with CD8+ regulatory T cells. Vectored Immunoprophylaxis as an Approach to Humoral Immunity Another approach to generating humoral immunity to HIV-1 is based not on induction of active B cell immunity but on vectored immunoprophylaxis (21, 22), In this strategy, a viral vector (e.g., adeno-associated virus, AAV) is used to infect or otherwise insert genes encoding intact potent broadly neutralizing antibodies into host cells ex vivo with subsequent implantation or in vivo. This scheme provides arguably passive immunity in that there is no administration of an immunogen related to HIV and no elicitation of an immune response involving host B lymphocytes, as usually defined. However, the production of antibodies is active in the host and no already synthesized antibodies are directly infused. Early studies in animal models have demonstrated proof of principle for using vectored immunoprophylaxis to confer robust protection to recipient animals challenged with significant doses of virulent HIV by clinically relevant routes (21, 22). Of course, applying vectored immunoprophylaxis on a mass scale as an alternative to standard vaccination also requires addressing so far unanswered questions. Who should receive the treatment and at what age? Is a single administration of the vector sufficient for long-lasting antibody production and protection? Are periodic administrations of the vector needed to maintain persistent protective immunity? Could HIV evolve so as to escape one or even multiple pbnAb generated via vectored immunoprophylaxis? If antibody produced by the vector produced undesirable side-effects, how could synthesis be abrogated in a timely manner? Will the genetic vectors persist in treated patients for periods and in ways that cause unwelcome side-effects? Can proponents of vectored immunoprophylaxis produce data that will assure the FDA that these safety concerns have been adequately addressed? Conclusion Due to a constellation of attributes including enormous genomic and antigenic sequence diversity, rapid evolution, and immunity-subverting structural features of key antigens associated with HIV-1, it is one of the most challenging pathogens vaccine developers have confronted to date. Technological advances in cloning Ig genes from individual human B lymphocytes, generating human monoclonal antibodies, and designing immunogens to express one or a limited number of epitopes have been extraordinary and rapid. These advances make plausible a vaccination scheme centered on the notion of using a series of related but non-identical immunogens to guide the process of B cell evolution through repeated rounds of somatic hypermutation leading to affinity maturation and acquisition of broadly neutralizing activity. However, numerous scientific and logistical challenges remain to be addressed before such a scheme could be implemented on a public health scale. Therefore, alternative strategies to generating protective immunity to HIV-1, such as vectored immunoprophylaxis or induction of regulatory responses intended to reduce activation of CD4+ T cells, remain worthy of thorough exploration. Conflict of Interest Statement The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

          Related collections

          Most cited references15

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

          Rational HIV immunogen design to target specific germline B cell receptors.

          Vaccine development to induce broadly neutralizing antibodies (bNAbs) against HIV-1 is a global health priority. Potent VRC01-class bNAbs against the CD4 binding site of HIV gp120 have been isolated from HIV-1-infected individuals; however, such bNAbs have not been induced by vaccination. Wild-type gp120 proteins lack detectable affinity for predicted germline precursors of VRC01-class bNAbs, making them poor immunogens to prime a VRC01-class response. We employed computation-guided, in vitro screening to engineer a germline-targeting gp120 outer domain immunogen that binds to multiple VRC01-class bNAbs and germline precursors, and elucidated germline binding crystallographically. When multimerized on nanoparticles, this immunogen (eOD-GT6) activates germline and mature VRC01-class B cells. Thus, eOD-GT6 nanoparticles have promise as a vaccine prime. In principle, germline-targeting strategies could be applied to other epitopes and pathogens.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Distribution and three-dimensional structure of AIDS virus envelope spikes.

            Envelope glycoprotein (Env) spikes on AIDS retroviruses initiate infection of host cells and are therefore targets for vaccine development. Though crystal structures for partial Env subunits are known, the structure and distribution of native Env spikes on virions is obscure. We applied cryoelectron microscopy tomography to define ultrastructural details of spikes. Virions of wild-type human immunodeficiency virus 1 (HIV-1) and a mutant simian immunodeficiency virus (SIV) had approximately 14 and approximately 73 spikes per particle, respectively, with some clustering of HIV-1 spikes. Three-dimensional averaging showed that the surface glycoprotein (gp120) 'head' of each subunit of the trimeric SIV spike contains a primary mass, with two secondary lobes. The transmembrane glycoprotein 'stalk' of each trimer is composed of three independent legs that project obliquely from the trimer head, tripod-like. Reconciling available atomic structures with the three-dimensional whole spike density map yields insights into the orientation of Env spike structural elements and possible structural bases of their functions.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The antigenic structure of the HIV gp120 envelope glycoprotein.

              The human immunodeficiency virus HIV-1 establishes persistent infections in humans which lead to acquired immunodeficiency syndrome (AIDS). The HIV-1 envelope glycoproteins, gp120 and gp41, are assembled into a trimeric complex that mediates virus entry into target cells. HIV-1 entry depends on the sequential interaction of the gp120 exterior envelope glycoprotein with the receptors on the cell, CD4 and members of the chemokine receptor family. The gp120 glycoprotein, which can be shed from the envelope complex, elicits both virus-neutralizing and non-neutralizing antibodies during natural infection. Antibodies that lack neutralizing activity are often directed against the gp120 regions that are occluded on the assembled trimer and which are exposed only upon shedding. Neutralizing antibodies, by contrast, must access the functional envelope glycoprotein complex and typically recognize conserved or variable epitopes near the receptor-binding regions. Here we describe the spatial organization of conserved neutralization epitopes on gp120, using epitope maps in conjunction with the X-ray crystal structure of a ternary complex that includes a gp120 core, CD4 and a neutralizing antibody. A large fraction of the predicted accessible surface of gp120 in the trimer is composed of variable, heavily glycosylated core and loop structures that surround the receptor-binding regions. Understanding the structural basis for the ability of HIV-1 to evade the humoral immune response should assist in the design of a vaccine.
                Bookmark

                Author and article information

                Contributors
                URI : http://frontiersin.org/people/u/111375
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                16 July 2014
                2014
                : 5
                : 335
                Affiliations
                [1] 1Department of Pathology, Case Western Reserve University , Cleveland, OH, USA
                Author notes

                Edited by: Marc H. V. Van Regenmortel, University of Strasbourg, France

                Reviewed by: Quentin James Sattentau, University of Oxford, UK; Marc H. V. Van Regenmortel, University of Strasbourg, France

                *Correspondence: nsg@ 123456case.edu

                This article was submitted to HIV and AIDS, a section of the journal Frontiers in Immunology.

                Article
                10.3389/fimmu.2014.00335
                4099939
                8ba672f2-407e-4a57-9c43-f1a10195e243
                Copyright © 2014 Greenspan.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 31 May 2014
                : 03 July 2014
                Page count
                Figures: 0, Tables: 0, Equations: 0, References: 22, Pages: 3, Words: 2888
                Categories
                Immunology
                Opinion Article

                Immunology
                genomic diversity,immune escape,broadly neutralizing antibody,non-neutralizing antibody,vaccine-guided b cell evolution,immunogen design,regulatory cd8+ t cells,vectored immunoprophylaxis

                Comments

                Comment on this article