Abstract
A prophylactic vaccine would be a powerful tool in the fight against HIV. Passive immunization of animals with broadly neutralizing antibodies (bnAbs) affords protection against viral challenge, and recent data from the Antibody Mediated Prevention clinical trials support the concept of bnAbs providing protection against HIV in humans, albeit only at broad and potent neutralizing antibody titers. Moreover, it is now clear that a successful vaccine will also need to induce bnAbs against multiple neutralizing epitopes on the HIV envelope (Env) glycoprotein. Here, we review recent clinical trials evaluating bnAb-based vaccines, and discuss key issues in the development of an HIV vaccine capable of targeting multiple Env neutralizing epitopes.
Introduction
Vaccines that induce broadly neutralizing antibodies (bnAbs) are a leading objective for HIV vaccine research. The recently published results of the Antibody Mediated Prevention (AMP) trials provided proof-of-concept that a bnAb-inducing vaccine could also protect people from infection [1]. However, the AMP trials also demonstrated an HIV vaccine that elicits only one specificity of bnAb would likely not be clinically effective. For an HIV vaccine to be successful, it will need to elicit bnAbs against several distinct epitopes on the envelope glycoprotein (Env). In this review, we discuss results of clinical trials of bnAbs for passive immunization and prophylaxis, and review strategies for design and delivery of bnAb-based vaccines.
HIV broadly neutralizing antibodies for prevention
BnAbs prevent HIV infection by binding to conserved epitopes on Env, thereby interfering with the viral machinery to enter target cells. Seven neutralizing sites of bnAbs have been identified on the Env gp120-gp41 trimer complex: sites on the first and second variable regions (V1/V2-glycan), sites on the third variable region (V3-glycan), the CD4 binding site (CD4bs), the gp120-gp41 interface, the membrane-proximal external region (MPER), the Env silent face, and the fusion domain [2–4] (Figure 1A). An eighth bnAb type, Fab-Dimerized Glycan-reactive (FDG) antibodies, represents a new class of bnAbs that bind only to glycans and can target multiple glycan sites [5]. Prototypical bnAbs for each neutralizing epitope, some of which are being tested in passive immunization studies for their capacity to protect people from infection, are shown in Figure 1B.
Figure 1:
Neutralizing epitopes on the HIV Envelope glycoprotein. (A) Neutralizing epitopes are mapped on a representative HIV Env trimer and shown in top view (left), side view (middle), and bottom view (right). BnAb epitopes are indicated by color: green, V3-glycan; red, V1/V2-glycan; blue, CD4bs; orange, fusion domain; magenta, Env silent face; cyan, gp41-gp120 interface. The position of the MPER region is shown in the side view and the first amino acid of the MPER peptide is marked with an asterisk in the bottom view. (B) Examples of bnAbs for each neutralizing epitope are listed and color-coded according to their epitope. An additional bnAb category, Fab-Dimerized Glycan-reactive (FDG) bnAbs have been recently described that bind to multiple Env glycans.
Can bnAbs protect humans from HIV infection?
BnAb infusions of humanized mice and non-human primates protect against HIV viral challenge [6–10]. A meta-analysis of rhesus macaque studies of bnAb administration followed by SHIV (chimeric simian-human immunodeficiency virus) challenge demonstrated that serum neutralization titer has a significant effect on infection risk and that viral protection by a single bnAb likely will require relatively high levels of neutralization at the time of exposure [11]. Administration of bnAbs to HIV-infected people transiently reduces viremia [12], confirming that bnAbs exert anti-viral effects in humans as well. The critical follow-up question has been do bnAbs protect people from HIV infection and, if so, what serum titer of bnAb is needed for protection?
The recently published results of the AMP trials [HIV Vaccine Trials Network (HVTN) 704/HIV Prevention Trials Network (HPTN) 085 and HVTN 703/HPTN 081] offer important insights [1]. In two parallel trials, at-risk cisgender men and transgender people (HVTN 704) or at-risk women (HVTN 703) were infused every eight weeks with a high or low dose of the CD4bs bnAb VRC01 or placebo for a total of ten infusions. VRC01 infusion did not provide a significant reduction in overall HIV transmission, but the frequency of transmission of VRC01-sensitive viruses was significantly lower in the treatment groups than in the placebo groups, with VRC01 infusion providing 75% protective efficacy against sensitive viruses [1]. This result provided proof-of-concept that passive immunization with bnAbs can protect people against viral acquisition. The lack of overall protection against HIV infection, however, highlights that any strategy for bnAb-based HIV prophylaxis will require consistently high titers of neutralizing antibodies against a wide breadth of HIV isolates.
The results of the AMP trials suggest that a mixture of bnAbs targeting different neutralizing epitopes will expand protection against various HIV isolates, and results of ex vivo neutralization studies using combinations of bnAbs targeting different neutralizing epitopes support this idea [13,14]. Clinical trials are underway to test whether infusion of two or three bnAbs targeting different epitopes indeed provide better protection (Table 1). An ongoing phase I clinical trial (NCT04173819) is evaluating the safety, tolerability, and pharmacokinetics of the combination of a CD4bs and a V3-glycan bnAb. Similar trials are testing different combinations of two or three antibodies that target different bnAb epitopes (NCT04212091 and NCT03928821). An alternative to using bnAb combinations is to engineer single antibodies bearing two or three different antigen binding sites, each of which binds a different bnAb epitope. Two phase I trials are underway to test the safety of bi- or tri-specific antibody approaches (NCT03875209 and NCT03705169).
Table 1:
Representative clinical trials of passive infusion of multiple bnAbs
Clinical trial | Concept | Trial Phase |
---|---|---|
NCT03571204 | Infusion of 3BNC117 (CD4bs) and 10-1074 (V3-glycan) bnAbs | Phase 1 |
NCT04173819 | Infusion of 3BNC117 (CD4bs) and 10-1074 (V3-glycan) bnAbs, i.v. vs. s.c. | Phase 1/2 |
NCT04212091 | Infusion of PGT121 (V3-glycan) and VRC07 (CD4bs) bnAbs | Phase 1 |
NCT03928821 | Infusion of combinations (two or three mAb) of PGT121 (V3-glycan), PGDM1400 (V1/V2-glycan, 10-1074 (V3-glycan), andVRC07 (CD4bs) | Phase 1 |
NCT03875209 | Bi-specific antibody injection 10E8.4/iMab (MPER + CD4) | Phase 1 |
NCT03705169 | Tri-specific antibody SAR441236 (CD4bs + V1/V2-glycan + MPER) | Phase 1 |
Can vaccines elicit bnAbs?
To date, bnAbs have only been induced by retroviral infection in HIV-infected individuals or SHIV-infected rhesus macaques [15,16]. Unfortunately, vaccination has not reliably induced potent bnAbs in animal models or human clinical trials. However, preclinical and clinical studies have informed our understanding of the immunological roadblocks in generating bnAbs through vaccination.
BnAbs exhibit unusual properties, including long heavy-chain complementarity-determining region (CDR) 3 loops and/or autoreactivity, which normally trigger immune tolerance mechanisms that prevent the development or activation of B cells bearing these receptors [17,18]. In addition, mature bnAbs contain a large number of somatic mutations and, in some cases, insertions or deletions in the immunoglobulin heavy and light chain genes, suggesting that prolonged periods of germinal center occupancy are needed for bnAb development [17]. Thus, the two major challenges for HIV vaccine development are: 1) design of “germline-targeting” immunogens that can activate rare and low-affinity bnAb precursor B cells, and 2) selection of bnAb precursor B cells to enter and stay in germinal centers in order to accumulate the mutations required for potency and breadth.
Two approaches have been used in the design of bnAb germline or unmutated common ancestor (UCA)-targeting immunogens: 1) well-folded, native Envs based on transmitted/founder viruses isolated from HIV-infected people who eventually generated bnAbs, and 2) proteins engineered to bind UCAs and mimic Env neutralizing epitopes. A key consideration in the development of a germline/UCA targeting immunogen is the affinity of the immunogen for B cell receptors (BCRs) expressed by naïve bnAb precursor B cells [19]. Transgenic mice, in which a subset of naïve B cells express the computationally inferred UCA precursor BCR for a certain bnAb lineage, are powerful tools for testing the capacity of novel immunogens to activate B cells with bnAb potential in vivo [20–23]. After immunization, serial analysis of the breadth of neutralization induced in plasma, the frequency of bnAb precursors induced in blood and lymphoid tissues, and the mutations induced in bnAb B cell lineage members provides insight as to how well a immunogen formulation is performing and predicts how well a vaccine-induced bnAb type or class will protect against viral strains. It is also important to determine if a candidate germline-targeting immunogen can activate not only inferred germline precursors in mice, but also authentic naïve B cells in humans. For example, B cells that bind the N332-GT2 and eOD-GT8 immunogens have been found in the naïve human B cell repertoire [22,23]. Isolating and characterizing reactive naïve human B cells will be critical for evaluating other germline targeting immunogens, although this process can be very challenging, since naïve germline precursors, particularly those for long HCDR3-bearing bnAbs, are rare and of low affinity [19].
Success with various germline or UCA targeting immunogens in pre-clinical studies has led to their evaluation in human clinical trials (Table 2). The eOD-GT8 60mer nanoparticle was recently tested in a phase I clinical trial (NCT03547245, IAVI G001), with early encouraging results [24]. In this trial, 48 HIV negative adults received two doses of eOD-GT8 or placebo two months apart. Most of the immunized individuals (97%) who received the eOD-GT8 60mer exhibited activated B cells with features of germline precursors of the VRC01 lineage.
Table 2:
Immunogens that target B cell germline precursors for bnAb lineages
Immunogen | Epitope | BnAb GL or UCA targeted | Clinical Trial |
---|---|---|---|
CH505 TF gp120 | CD4bs | CH103 | HVTN 115 (NCT03220724) |
CH848 10.17DT NP | V3-glycan | DH270.6 | HVTN 3XX |
BG505 MD39.3 Trimer | V3-glycan | PGT121 | HVTN 302 |
eOD-GT8 | CD4bs | VRC01 | IAVI G001 (NCT03547245) |
BG505 SOSIP.GT1.1 gp140 trimer | CD4bs | VRC01 | IAVI C101 (NCT04224701) |
CH505 TF SOSIP Trimer (Clade A BG505 backbone) | CD4bs | CH103 | HVTN 300 |
426c Core NP | CD4bs | VRC01 | HVTN 301 |
MPER-656 peptide-liposome | MPER | 2F5, 10E8, 4E10, DH511 | HVTN 133 (NCT03934541) |
Design of bnAb boosting immunogens
The results of germline/UCA targeting immunogens in animal studies and early clinical trials have raised optimism that bnAb precursors are present and functional in the naïve B cell pool. However, germline/UCA targeting immunogens alone have yet to induce the degree of somatic mutation in bnAb lineages needed for neutralization breadth. Thus, there is considerable focus on the use of sequential boosting immunogens that mature bnAb lineages in germinal centers [17].
The absolute number of mutations required to generate bnAbs from germline precursors represents a substantial hurdle in itself, but research in recent years has revealed additional challenges that must be overcome for a boosting immunogen to advance a bnAb lineage. Most bnAbs require somatic mutations in the framework regions of their immunoglobulin genes, which facilitates neutralization breadth by conferring the antibody with greater flexibility in antigen binding sites [25]. However, framework region mutations are less tolerated than mutations in antigen-contacting CDR loops, which may negatively impact B-cell fitness in germinal centers [25]. In addition, bnAbs are enriched for mutations in “cold” spots, sequences of immunoglobulin genes that are seldom subject to somatic mutation, and thus represent improbable events [26]. Lastly, approximately 40% of bnAbs require in-frame insertions or deletions, which occur only rarely during somatic hypermutation [27]. Thus, an effective boosting immunogen will need to bind BCRs on the mutated descendants of bnAb germline precursors, and then promote the selection of B cells expressing BCRs that were produced through rare events in the somatic hypermutation process. Nonetheless, the feasibility of the sequential immunization strategy has been demonstrated in mice, where immunization of bnAb precursor transgenic mice with germline targeting immunogens followed by boosts with heterologous immunogens results in greater neutralization breadth than repeated immunization with germline targeting immunogens alone [28,29].
An important consideration in the design of Env immunogens is the presence of immunodominant non-neutralizing epitopes that can take B cell lineages “off track.” These epitopes are normally occluded on the Env trimer but are exposed upon receptor CD4 binding or upon disassembly of Env trimers into monomers. To ensure that Env immunogens are maintained in a well-folded conformation following administration, multiple mutations have been integrated into immunogens that stabilize the Env trimer and minimize the exposure of undesired off-target epitopes [30–32].
Designing an HIV vaccine capable of inducing multiple specificities of bnAbs
Can germline targeting and sequential immunization approaches be adapted for a multi-bnAb vaccine? One might envision a regimen of immunogen cocktails, with a priming cocktail capable of activating bnAb germline precursors for various neutralizing epitopes, and subsequent boosting cocktails containing the next immunogens in each sequence that mature different bnAb lineages (Figure 2). With the induction of bnAbs against multiple epitopes would come protection against a broad spectrum of HIV strains (Figure 2).
Figure 2:
The sequential immunization approach for a single vs. multi-bnAb HIV vaccine. For a bnAb-inducing vaccine that targets a single neutralizing epitope (top), a germline (GL) targeting immunogen is administered for immunization #1 to activate bnAb precursors. Subsequent boosting immunizations rely on modified immunogens that favor the acquisition of mutations in immunoglobulin genes that increase neutralizing breadth. Induction of bnAbs against a single neutralizing epitope, however, will offer protection only against a subset of sensitive viruses. In contrast, a multi-bnAb vaccine (bottom) would rely on multiple immunogens that induce antibody lineages against different neutralizing epitopes. With multiple bnAbs induced, vaccine recipients will be protected against larger fraction of viruses.
An important consideration for a multi-bnAb vaccine will be the method for delivering several immunogens in one injection. Strategies to deliver multiple immunogens can be categorized into three approaches: 1) mixing of individual immunogens; 2) design of chimeric immunogens featuring targeting motifs for different neutralizing epitopes; and 3) display of different immunogens on the surface of a nanoparticle. The mixing of different immunogens to elicit antibodies against a variety of pathogen variants has been exploited for a variety of vaccines, including the pneumococcal vaccine, in which polysaccharides of different bacterial serotypes are combined to elicit an array of serotype-specific antibodies [33]. The efficacy of multivalent pneumococcal vaccines derives largely from the highly immunogenic nature of the bacterial polysaccharides, to the point that antibodies can be generated without T cell help. Thus, mixing of germline targeting or boosting immunogens for HIV may represent the simplest approach, but may lack the necessary immunogenicity to drive bnAb maturation in germinal centers. Moreover, a mixed HIV vaccine may suffer at the hands of pragmatism; the hypothetical immunization strategy shown in Figure 2, comprising a four-injection regimen with four bnAb-inducing immunogens per immunization, represents sixteen individual immunogens to undergo regulatory review and manufacture.
A chimeric immunogen approach, in which targeting motifs for different neutralizing epitopes are incorporated onto a common backbone, can overcome the “too many immunogens” issue. Medina-Ramirez et al. demonstrated in transgenic mice that a two-epitope germline targeting protein was able to bind and activate germline precursors of two bnAb lineages, VRC01 and PGT121 [34]. Similarly, Saunders et al. demonstrated that a native Env-based CD4bs germline targeting immunogen was capable of eliciting V1/V2-glycan targeted neutralizing antibodies [35]. It is unclear, however, how many neutralizing epitopes can be incorporated into a single immunogen, and whether the correct sequential epitopes can be readily incorporated into the same molecular backbone.
Multimerizing Env immunogens on nanoparticles has become an important strategy to overcome the lack of Env immunogenicity, and it may have the added bonus of offering a platform for delivering different HIV immunogens on the same molecule. There is growing evidence that viral immunogens arrayed on a particle enhance B cell activation and induce higher antibody titers than soluble forms of the same immunogen [21,36–38]. Particle-based strategies include display on a variety of structures such as virus-like particles, liposomes, lipid nanocapsules, silica nanoparticles, and ferritin nanoparticles. The clinical trial of eOD-GT8 60mer, for example, used a multimerization strategy in which an average of 60 copies of the eOD-GT8 immunogen were arrayed on a bacterial lumazine synthase-based particle [39]. The HVTN 133 trial studies an MPER neutralizing epitope embedded in a liposome to mimic epitope presentation on a viral membrane, and in macaques initiated an MPER bnAb lineage [40]. Brouwer et al. engineered a self-assembling nanoparticle that is capable of displaying a variety of different Envs; in their original report, homogenous nanoparticles of Envs were tested, but platforms of this type may allow for the display of different immunogens on the same nanoparticle [41]. Indeed, recent studies have revealed the feasibility of displaying multiple immunogens on the same particle. Cohen et al. generated nanoparticles displaying Spike proteins from eight different coronavirus species as a strategy to elicit multiple neutralizing antibody lineages [42]. Most recently, a scaffolded nanoparticle quadrivalent influenza vaccine decorated with four different hemagglutinin molecules has been reported that induced broad influenza protection [43].
Multiple protein-based immunogens, however, may prove logistically impractical or prohibitively expensive for a multi-bnAb approach. Thus, an alternative strategy could employ nucleoside-modified mRNAs to encode multiple HIV immunogens. Modified mRNA vaccines offer many features that are amenable to a multi-bnAb, sequential immunization approach [44,45]. Modified mRNAs are simpler and faster to manufacture than traditional protein immunogens and, importantly, are potent inducers of T follicular helper cells and B cell germinal centers [46]. Moreover, multiple immunogens can be encoded as modified mRNAs and delivered in lipid nanoparticles: a modified mRNA vaccine encoding multiple influenza antigens protected mice against a panel of different influenza viruses [47]. Results from pre-clinical studies for modified mRNA-based, single bnAb HIV vaccines are ongoing [48,49], but studies with multiple co-administered modified mRNA-encoded immunogens are needed.
Conclusion
Considerable knowledge has been gained over the past three decades about the barriers to HIV vaccine-induced bnAb generation. The B cell germline precursors for bnAb lineages are rare, some are disfavored by immune tolerance mechanisms, and the path from germline precursors to bnAbs is torturous, requiring rare somatic hypermutations. Nonetheless, pre-clinical studies in animals suggest that optimal combination of immunogens may be able to promote bnAb development.
However, the results of the AMP trial were clear: inducing bnAbs can protect against susceptible HIV strains but the titers needed to protect will be relatively high, and it will require antibodies to multiple epitopes to protect against viral diversity. If a bnAb against a single neutralizing epitope can be guided through full maturation, the next challenge will be to design vaccines that elicit bnAbs against multiple neutralizing epitopes to maximize coverage against viral variants. Challenges for a multi-bnAb vaccine will likely involve the format for delivering multiple immunogens in a manner that elicits optimal responses from B cell germline precursors and intermediates for different bnAb lineages. With the development of chimeric immunogens, novel nanoparticle platforms, and modified mRNA-based vaccines, there is a growing toolbox for the delivery of a multi-bnAb HIV vaccine.
Acknowledgments
We are grateful to Dr. Ashley Bennet for her assistance in developing Figure 1. This work was supported by the NIAID, NIH UM1 Consortium for HIV/AIDS Vaccine Development (CHAVD) [Grant UM1AI144371].
Footnotes
Declaration of interest: BFH as patent applications on some of the concepts and immunogens discussed in this paper.
References
- 1.**.Corey L, Gilbert PB, Juraska M, Montefiori DC, Morris L, Karuna ST, Edupuganti S, Mgodi NM, deCamp AC, Rudnicki E, et al. Two Randomized Trials of Neutralizing Antibodies to Prevent HIV-1 Acquisition. N Engl J Med 2021, 384:1003–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]; Results from the Antibody Mediated Prevention (AMP) clinical trials, where infusion of CD4bs bnAb VRC01 in high-risk individuals prevented infection from VRC01 sensitive HIV strains.
- 2.**.Haynes BF, Burton DR, Mascola JR: Multiple roles for HIV broadly neutralizing antibodies. Sci Transl Med 2019, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]; A recent insightful review article covers the progress of using HIV broadly neutralizing antibodies in passive immunizations and strategies of developing vaccines that elicit broadly neutralizing antibodies.
- 3.Steichen JM, Kulp DW, Tokatlian T, Escolano A, Dosenovic P, Stanfield RL, McCoy LE, Ozorowski G, Hu X, Kalyuzhniy O, et al. HIV Vaccine Design to Target Germline Precursors of Glycan-Dependent Broadly Neutralizing Antibodies. Immunity 2016, 45:483–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.**.Kwong PD, Mascola JR: HIV-1 Vaccines Based on Antibody Identification, B Cell Ontogeny, and Epitope Structure. Immunity 2018, 48:855–871. [DOI] [PubMed] [Google Scholar]; This comprehensive review covers the development and molecular characteristics of HIV broadly neutralizing antibodies, as well as multiple strategies to design bnAb-inducing HIV vaccines.
- 5.Williams WB, Meyerhoff RR, Edwards RJ, Li H, Manne K, Nicely NI, Henderson R, Zhou Y, Janowska K, Mansouri K, et al. Fab-dimerized glycan-reactive antibodies are a structural category of natural antibodies. Cell 2021, 184:2955–2972 e2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Shingai M, Donau OK, Plishka RJ, Buckler-White A, Mascola JR, Nabel GJ, Nason MC, Montefiori D, Moldt B, Poignard P, et al. Passive transfer of modest titers of potent and broadly neutralizing anti-HIV monoclonal antibodies block SHIV infection in macaques. Journal of Experimental Medicine 2014, 211:2061–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Saunders KO, Pegu A, Georgiev IS, Zeng M, Joyce MG, Yang ZY, Ko SY, Chen XJ, Schmidt SD, Haase AT, et al. Sustained Delivery of a Broadly Neutralizing Antibody in Nonhuman Primates Confers Long-Term Protection against Simian/Human Immunodeficiency Virus Infection. Journal of Virology 2015, 89:5895–5903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Pegu A, Yang ZY, Boyington JC, Wu L, Ko SY, Schmidt SD, Mckee K, Kong WP, Shi W, Chen XJ, et al. Neutralizing antibodies to HIV-1 envelope protect more effectively in vivo than those to the CD4 receptor. Science Translational Medicine 2014, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Klein F, Halper-Stromberg A, Horwitz JA, Gruell H, Scheid JF, Bournazos S, Mouquet H, Spatz LA, Diskin R, Abadir A, et al. HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature 2012, 492:118–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gautam R, Nishimura Y, Pegu A, Nason MC, Klein F, Gazumyan A, Golijanin J, Buckler-White A, Sadjadpour R, Wang K, et al. A single injection of anti-HIV-1 antibodies protects against repeated SHIV challenges. Nature 2016, 533:105–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pegu A, Borate B, Huang Y, Pauthner MG, Hessell AJ, Julg B, Doria-Rose NA, Schmidt SD, Carpp LN, Cully MD, et al. A Meta-analysis of Passive Immunization Studies Shows that Serum-Neutralizing Antibody Titer Associates with Protection against SHIV Challenge. Cell Host Microbe 2019, 26:336–346 e333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Caskey M, Klein F, Lorenzi JC, Seaman MS, West AP, Buckley N Jr, Kremer G, Nogueira L, Braunschweig M, Scheid JF, et al. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 2015, 522:487–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kong R, Louder MK, Wagh K, Bailer RT, deCamp A, Greene K, Gao H, Taft JD, Gazumyan A, Liu C, et al. Improving neutralization potency and breadth by combining broadly reactive HIV-1 antibodies targeting major neutralization epitopes. J Virol 2015, 89:2659–2671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Berendam SJ, Styles TM, Morgan-Asiedu PK, Tenney D, Kumar A, Obregon-Perko V, Bar KJ, Saunders KO, Santra S, De Paris K, et al. Systematic Assessment of Antiviral Potency, Breadth, and Synergy of Triple Broadly Neutralizing Antibody Combinations against Simian-Human Immunodeficiency Viruses. J Virol 2021, 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Liao HX, Lynch R, Zhou T, Gao F, Alam SM, Boyd SD, Fire AZ, Roskin KM, Schramm CA, Zhang Z, et al. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 2013, 496:469–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Roark RS, Li H, Williams WB, Chug H, Mason RD, Gorman J, Wang S, Lee FH, Rando J, Bonsignori M, et al. Recapitulation of HIV-1 Env-antibody coevolution in macaques leading to neutralization breadth. Science 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Haynes BF, Kelsoe G, Harrison SC, Kepler TB: B-cell-lineage immunogen design in vaccine development with HIV-1 as a case study. Nat Biotechnol 2012, 30:423–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shi B, Ma L, He X, Wang X, Wang P, Zhou L, Yao X: Comparative analysis of human and mouse immunoglobulin variable heavy regions from IMGT/LIGM-DB with IMGT/HighV-QUEST. Theor Biol Med Model 2014, 11:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Havenar-Daughton C, Abbott RK, Schief WR, Crotty S: When designing vaccines, consider the starting material: the human B cell repertoire. Current Opinion in Immunology 2018, 53:209–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.*.Steichen JM, Lin YC, Havenar-Daughton C, Pecetta S, Ozorowski G, Willis JR, Toy L, Sok D, Liguori A, Kratochvil S, et al. A generalized HIV vaccine design strategy for priming of broadly neutralizing antibody responses. Science 2019, 366:1216-+. [DOI] [PMC free article] [PubMed] [Google Scholar]; Using bnAb BG18 as a test case, the author proposed a strategy that uses ultra-deep human antibody sequencing data to identify bnAb precursors and engineer germline targeting priming immunogens. The authors believe this strategy could be applied to many HIV bnAbs.
- 21.*.Saunders KO, Wiehe K, Tian M, Acharya P, Bradley T, Alam SM, Go EP, Scearce R, Sutherland L, Henderson R, et al. Targeted selection of HIV-specific antibody mutations by engineering B cell maturation. Science 2019, 366:1215-+. [DOI] [PMC free article] [PubMed] [Google Scholar]; The authors designed and tested two germline targeting immunogens for V3-glycan bnAb lineage DH270.6 and CD4bs bnAb lineage CH235. These two immunogens elicited antibodies with functional bnAb mutations in germline precursor knock-in mice and rhesus macaques.
- 22.Jardine JG, Ota T, Sok D, Pauthner M, Kulp DW, Kalyuzhniy O, Skog PD, Thinnes TC, Bhullar D, Briney B, et al. Priming a broadly neutralizing antibody response to HIV-1 using a germline-targeting immunogen. Science 2015, 349:156–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.**.Huang DL, Abbott RK, Havenar-Daughton C, Skog PD, Al-Kolla R, Groschel B, Blane TR, Menis S, Tran JT, Thinnes TC, et al. B cells expressing authentic naive human VRC01-class BCRs can be recruited to germinal centers and affinity mature in multiple independent mouse models. Proceedings of the National Academy of Sciences of the United States of America 2020, 117:22920–22931. [DOI] [PMC free article] [PubMed] [Google Scholar]; Using adoptive transfer mouse model, the author demonstrated that B cells expressing authentic naive human VRC01-class BCR at rare human physiologically-relative frequencies were activated, recruited into germinal centers, and formed memory B cells by eOD-GT8 60mer nanoparticles.
- 24.Venkatesan P: Preliminary phase 1 results from an HIV vaccine candidate trial. Lancet Microbe 2021, 2:E95–E95. [DOI] [PubMed] [Google Scholar]
- 25.Klein F, Diskin R, Scheid JF, Gaebler C, Mouquet H, Georgiev IS, Pancera M, Zhou T, Incesu RB, Fu BZ, et al. Somatic mutations of the immunoglobulin framework are generally required for broad and potent HIV-1 neutralization. Cell 2013, 153:126–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wiehe K, Bradley T, Meyerhoff RR, Hart C, Williams WB, Easterhoff D, Faison WJ, Kepler TB, Saunders KO, Alam SM, et al. Functional Relevance of Improbable Antibody Mutations for HIV Broadly Neutralizing Antibody Development. Cell Host & Microbe 2018, 23:759-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kepler TB, Liao HX, Alam SM, Bhaskarabhatla R, Zhang R, Yandava C, Stewart S, Anasti K, Kelsoe G, Parks R, et al. Immunoglobulin gene insertions and deletions in the affinity maturation of HIV-1 broadly reactive neutralizing antibodies. Cell Host Microbe 2014, 16:304–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Escolano A, Steichen JM, Dosenovic P, Kulp DW, Golijanin J, Sok D, Freund NT, Gitlin AD, Oliveira T, Araki T, et al. Sequential Immunization Elicits Broadly Neutralizing Anti-HIV-1 Antibodies in Ig Knockin Mice. Cell 2016, 166:1445–1458 e1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chen X, Zhou T, Schmidt SD, Duan H, Cheng C, Chuang GY, Gu Y, Louder MK, Lin BC, Shen CH, et al. Vaccination induces maturation in a mouse model of diverse unmutated VRC01-class precursors to HIV-neutralizing antibodies with >50% breadth. Immunity 2021, 54:324–339 e328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kwon YD, Pancera M, Acharya P, Georgiev IS, Crooks ET, Gorman J, Joyce MG, Guttman M, Ma X, Narpala S, et al. Crystal structure, conformational fixation and entry-related interactions of mature ligand-free HIV-1 Env. Nat Struct Mol Biol 2015, 22:522–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Medina-Ramirez M, Sanders RW, Sattentau QJ: Stabilized HIV-1 envelope glycoprotein trimers for vaccine use. Current Opinion in Hiv and Aids 2017, 12:241–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhang P, Gorman J, Geng H, Liu QB, Lin Y, Tsybovsky Y, Go EP, Dey B, Andine T, Kwon A, et al. Interdomain Stabilization Impairs CD4 Binding and Improves Immunogenicity of the HIV-1 Envelope Trimer. Cell Host & Microbe 2018, 23:832-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Daniels CC, Rogers PD, Shelton CM: A Review of Pneumococcal Vaccines: Current Polysaccharide Vaccine Recommendations and Future Protein Antigens. J Pediatr Pharmacol Ther 2016, 21:27–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Medina-Ramirez M, Garces F, Escolano A, Skog P, de Taeye SW, Del Moral-Sanchez I, McGuire AT, Yasmeen A, Behrens AJ, Ozorowski G, et al. Design and crystal structure of a native-like HIV-1 envelope trimer that engages multiple broadly neutralizing antibody precursors in vivo. J Exp Med 2017, 214:2573–2590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Saunders KO, Verkoczy LK, Jiang C, Zhang J, Parks R, Chen H, Housman M, Bouton-Verville H, Shen X, Trama AM, et al. Vaccine Induction of Heterologous Tier 2 HIV-1 Neutralizing Antibodies in Animal Models. Cell Rep 2017, 21:3681–3690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Asbach B, Wagner R: Particle-based delivery of the HIV envelope protein. Curr Opin HIV AIDS 2017, 12:265–271. [DOI] [PubMed] [Google Scholar]
- 37.Tokatlian T, Read BJ, Jones CA, Kulp DW, Menis S, Chang JYH, Steichen JM, Kumari S, Allen JD, Dane EL, et al. Innate immune recognition of glycans targets HIV nanoparticle immunogens to germinal centers. Science 2019, 363:649-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.*.Kato Y, Abbott RK, Freeman BL, Haupt S, Groschel B, Silva M, Menis S, Irvine DJ, Schief WR, Crotty S: Multifaceted Effects of Antigen Valency on B Cell Response Composition and Differentiation In Vivo. Immunity 2020, 53:548-+. [DOI] [PMC free article] [PubMed] [Google Scholar]; Using a series of HIV immunogens that vary in affinity and valency, the authors showed that high valency immunogens activate B cells rapidly with little affinity discrimination, while low valency immunogens tend to activate higher affinity B cells.
- 39.Jardine J, Julien JP, Menis S, Ota T, Kalyuzhniy O, McGuire A, Sok D, Huang PS, MacPherson S, Jones M, et al. Rational HIV immunogen design to target specific germline B cell receptors. Science 2013, 340:711–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhang RJ, Verkoczy L, Wiehe K, Alam SM, Nicely NI, Santra S, Bradley T, Pemble CW, Zhang JS, Gao F, et al. Initiation of immune tolerance-controlled HIV gp41 neutralizing B cell lineages. Science Translational Medicine 2016, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.*.Brouwer PJM, Antanasijevic A, Berndsen Z, Yasmeen A, Fiala B, Bijl TP, Bontjer I, Bale JB, Sheffler W, Allen JD, et al. Enhancing and shaping the immunogenicity of native-like HIV-1 envelope trimers with a two-component protein nanoparticle. Nature Communications 2019, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]; Design and testing of a two-component protein nanoparticle displaying 20 Env trimers which showed better immunogenicity than conventional one-component nanoparticles.
- 42.Cohen AA, Gnanapragasam PNP, Lee YE, Hoffman PR, Ou S, Kakutani LM, Keeffe JR, Wu HJ, Howarth M, West AP, et al. Mosaic nanoparticles elicit cross-reactive immune responses to zoonotic coronaviruses in mice. Science 2021, 371:735–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.*.Boyoglu-Barnum S, Ellis D, Gillespie RA, Hutchinson GB, Park YJ, Moin SM, Acton OJ, Ravichandran R, Murphy M, Pettie D, et al. Quadrivalent influenza nanoparticle vaccines induce broad protection. Nature 2021, 592:623-+. [DOI] [PMC free article] [PubMed] [Google Scholar]; Displaying 20 hemagglutinin trimers from the licensed quadrivalent influenza vaccines on a two-component nanoparticle induced broadly protective antibody responses.
- 44.Mu Z, Haynes BF, Cain DW: HIV mRNA Vaccines-Progress and Future Paths. Vaccines (Basel) 2021, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pardi N, Hogan MJ, Porter FW, Weissman D: mRNA vaccines - a new era in vaccinology. Nature Reviews Drug Discovery 2018, 17:261–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pardi N, Hogan MJ, Naradikian MS, Parkhouse K, Cain DW, Jones L, Moody MA, Verkerke HP, Myles A, Willis E, et al. Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J Exp Med 2018, 215:1571–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Freyn AW, da Silva JR, Rosado VC, Bliss CM, Pine M, Mui BL, Tam YK, Madden TD, Ferreira LCD, Weissman D, et al. A Multi-Targeting, Nucleoside-Modified mRNA Influenza Virus Vaccine Provides Broad Protection in Mice. Molecular Therapy 2020, 28:1569–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Pardi N, LaBranche CC, Ferrari G, Cain DW, Tombacz I, Parks RJ, Muramatsu H, Mui BL, Tam YK, Kariko K, et al. Characterization of HIV-1 Nucleoside-Modified mRNA Vaccines in Rabbits and Rhesus Macaques. Mol Ther Nucleic Acids 2019, 15:36–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Saunders KO, Pardi N, Parks R, Santra S, Mu ZK, Sutherland L, Scearce R, Barr M, Eaton A, Hernandez G, et al. Lipid nanoparticle encapsulated nucleoside-modified mRNA vaccines elicit polyfunctional HIV-1 antibodies comparable to proteins in nonhuman primates. Npj Vaccines 2021, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]