Discovery medicine – the HVTN's iterative approach to developing an HIV-1 broadly neutralizing vaccine

INTRODUCTION

Despite over 20 years of intensive effort, developing an HIV-1 vaccine has proven to be one of the greatest challenges in vaccinology. Only one vaccine regimen has shown modest efficacy [1], and the efficacy has so far not been reproducible elsewhere [2]. Catalyzed by advancements in immunogen design, sequencing, and structural analyses, the field's attention has shifted to development of a multifocal broadly neutralizing antibody (bnAb) vaccine, and there is a growing pipeline of candidate products. The HIV Vaccine Trials Network (HVTN) has developed the Discovery Medicine Program to evaluate bnAb-inducing vaccine candidates efficiently and systematically, emphasizing streamlined processes for rapid vaccine design iteration.

Approximately 30% of persons living with chronic HIV-1 infection develop bnAbs, often with unique features such as long heavy chain complementary determining regions (HCDR3 s), high levels of somatic hypermutation (SHM), insertions and deletions, and rare and improbable mutations [3–18]. Six known epitope regions on the HIV-1 envelope (Env) are susceptible to bnAb development, some with very high breadth and potency: CD4+-binding site (CD4bs), V2 apex, V3 glycan, gp120-gp41 interface, fusion peptide, and the membrane proximal external region (MPER) [1,3,19,20]. However, their naive B cell precursors can be extremely rare. Fortunately, the recent landmark eOD-GT8 study showed that, with the right immunogen, rare germline precursors [21,22] can be activated to induce first-step VRC01-class bnAbs in nearly all trial participants [23▪▪]. In addition, the Antibody Mediated Prevention (AMP) trials showed that passive infusion of a potent antibody like VRC01 is protective against HIV-1 strains that are neutralization-sensitive to VRC01 [24▪▪]. These results, together with the extraordinary success of the COVID-19 mRNA vaccines and improvements in immunogen design, have led to renewed enthusiasm for the development of HIV-1 neutralizing vaccines. 

FB1Box 1:

no caption available

THE SCIENCE BEHIND BNAB VACCINES Induction of bnAbs

Upon antigen exposure, a diverse pool of naive B cells expressing unmutated B-cell receptors (BCRs) become activated, migrate to lymph nodes, and form productive germinal centers composed of groups of B cells with distinct V(D)J rearrangements [25]. Precursor frequency and affinity with the immunogen determine the competitive fitness of activated B cells in germinal centers [26] and ultimately drive quality and quantity of memory B cell (MBC) and plasma cell outputs [25,27,28]. The goal of a germline or lineage-targeting vaccine is to activate a pool of naive, precursor B cells which, with continued maturation, can ultimately yield bnAbs of high neutralization potency and breadth. This process is mimicked in sequential vaccine design processes known as ‘priming’, ‘shepherding’ (or ‘shaping’), and ‘polishing’ [19,29]. These precursor B cells do not typically bind wild-type HIV-1 Env, nor do they neutralize the wild-type virus; therefore, the priming step must activate an unknown naive B cell whereafter boosting agents must bind unknown intermediate stage BCRs. In this way, subsequent heterologous boosting agents pull BCR development down a maturation pathway gradient, encouraging expression of rare key mutations that confer increasing affinity to the HIV trimer region that results in a fully developed bnAb. The B cell lineage for bnAbs, even those directed at the same region, are not uniform – for example, some people have multiple bnAbs directed at the same epitope such as the CD4bs. Whether there is an in vivo difference in the neutralizing efficiency of such bnAbs is unclear and will require further study, but the presence of multiple lineages provides optimism that the ability to prime and shepherd B cells into undergoing the uncommon mutations required of a bnAb can be achieved.

Immunogen design and sequential vaccination strategy

Three major approaches for bnAb germline initiation are being tested in the HVTN Discovery Medicine Program (Fig. 1). The first, ‘structure-based immunogen design’, uses a retrograde approach to immunogen development [8,11,30,31]. The priming antigen corresponding to selected bnAb germlines is computationally designed based on iterative affinity improvements with a pool of bnAb inferred germline (iGL) precursor antibodies. Successive Env immunogens are tested for binding to ultimately select a priming immunogen that can activate as many iGLs as possible [21,32–35,36▪▪].

F1FIGURE 1: HVTN Discovery Medicine Program. There are 22 trials in the HVTN pipeline targeting five epitopes using several platforms and adjuvants (top). Key innovations include the use of chimeric trimers, escalating dose priming, and comparisons between protein, protein nanoparticle, and mRNA immunogen platforms. Immunogen design is supported by rigorous preclinical data including use of KI mouse and NHP models (bottom). Adapted in part from [1].

The second approach, ‘mutation guided immunogen design’, uses an anterograde strategy for epitope-targeting prime and boost development [37,38]. HIV-1 envelopes thought to activate undifferentiated common ancestor (UCA) B cells are modified for adequate UCA activation, while simultaneously having higher affinity for the next intermediate stage B cell in the bnAb developmental pathway. The UCA/Env pairs are selected from individuals with chronic HIV-1 infection that develop potent and broad bnAbs. This affinity gradient-driven approach seeks to select rare but essential mutations necessary for maturation. Both programs use epitope-specific bnAbs with high potency and high breadth to model immunogen design. Preclinical testing includes knockin mouse models expressing human bnAb variable heavy and variable light chains [39▪], followed often by testing in nonhuman primates (rhesus macaques).

The third strategy, a ‘germline/lineage agnostic immunofocusing approach’, uses conserved Env peptides from the fusion peptide or MPER regions to induce broad, multigermline responses. Multiple distinct classes of fusion peptide bnAbs have been induced by fusion peptide immunogens in animal models [40,41]. The MPER peptide has both proximal and distal epitope targets, and an immunogen targeting the proximal region has induced Tier 2 antiviral responses in HVTN 133 (NCT03934541) [42].

A final interesting approach in advanced development is to couple immunogens designed with one of the above strategies to the Fc portion of dendritic cell targeting antibodies to better amplify the germinal center reaction for specific antigens [43,44].

CLINICAL TRIAL DESIGN

Discovery Medicine clinical trials are uniquely shaped by the overarching program goals. In contrast to more traditional clinical trials, they focus on gaining knowledge to facilitate iterative advancement in vaccine development. Trial sample size usually falls in the range of 10–20 participants in each study arm, with an emphasis on rapid assessment of safety and immunogenicity [45]. While placebos are sometimes included [46], they are often not necessary as the focus is on deep individual-level analysis to determine vaccine effects compared to baseline. An important feature of these trials is the ability to expand study scope based on early immune response data, collected typically after the second or third vaccination, to allow sufficient time for the emerging of bnAb precursors. This is accomplished by a Part B or Part C extension of the trial, with additional participants enrolled for the assessment of related regimens.

KEY DISCOVERY MEDICINE TRIALS HVTN 302: mRNA BG505 MD39.3 trimers

BG505 MD39.3 is a modified Clade A BG505 SOSIP, which includes stabilizing and glycan hole masking mutations [34]. HVTN 302 (NCT05217641) tests three versions of BG505 MD39.3, including a soluble gp140 trimer, a membrane-bound gp151 trimer, and a membrane-bound gp151 with a CD4bs knock-out mutation that confers a 1000-fold reduced avidity for CD4+ T cells [47]. This trial includes two doses, 100 and 250 μg of RNA, the higher of which is near the upper limits tested with current COVID-19 mRNA vaccines [48]. This study will help determine the relative advantages of mRNA-delivered soluble trimers vs. membrane-bound trimers. The first hypothesis is that membrane-bound bound trimers will present a more favorable orientation for an immune-focusing strategy, masking the immunogenic base, and therefore limiting the nonneutralizing, base-targeting B-cell response and improving B-cell responses targeting nonbase bnAb epitopes. Second, the CD4bs knockout immunogen reduces steric changes that occur during CD4+ binding that expose nonproductive immune sites and maintain bnAb epitopes. Results of this trial will improve understanding of the magnitude and quality of immune responses to mRNA-encoded HIV-1 trimers and inform further iterative immunogen development.

HVTN 301: 426c.Mod.Core-C4b nanoparticle – a CD4bs VRC01-class immunogen

HVTN 301 (NCT05471076) uses a VRC01-class-germline targeting immunogen known as the 426c.Mod.Core-C4b, a self-assembling 7-mer nanoparticle, adjuvanted with 3M-052-AF and alum. VRC01-class of bnAbs are desirable because they have been isolated from many individuals across the globe and despite 30% amino acid divergence and use of different angles of approach, they retain high potency and breadth [16,49–51] attributed to their CD4+ mimicking strategy. Several VRC01 targeting immunogens are in clinical trials, including the outer domain eOD-GT8 NP [23▪▪,32,52,53] and a trimer approach with BG505 GT1.1 [54]. The 426c.Mod.Core-C4b immunogen consists of the core inner and outer domains of the HIV-1 Clade C 426c transmitted founder viral envelope, including removal of the V1-V3 loops and several glycans, around the CD4bs, and importantly N276, required for germline binding [33,55–57].

The study goals are to assess whether naive B cells expressing VRC01-like B-cell receptors proliferate following immunization with a germline-targeting recombinant envelope; whether escalating dose priming [58–60], a strategy where the complete priming dose is divided up into multiple smaller escalating doses and delivered over two weeks, improves B cell activation over standard bolus dosing; and whether a high vs. low-dose boost improves VRC01-class B cell affinity maturation.

HVTN 144: N332-GT5 – testing a V3 glycan prime and generalizable HCDR3 bnAb-dependent design approach

HVTN 144 tests the N332-GT5 gp140 immunogen, a Clade A BG505 SOSIP trimer derivative, designed to induce V3 glycan BG19-class bnAb precursors, and is the first study targeting this epitope. This will also be the first human test of a generalizable strategy to prime HCDR3-dominant binding bnAbs through use of a computational engineering approach outlined by Steichen et al.[36▪▪]. While preclinical results were promising [34,35], if successful in humans it will validate the same approach for other HCDR3-dominant bnAb approaches targeting the V2 Apex, MPER, and FP regions. In addition, the trial will test a promising new adjuvant, saponin/monophosphoryl lipid A (MPLA) nanoparticle (SMNP), wherein saponin matrix technology is combined with the TLR4 agonist MPLA [61]. It will also test two dosing strategies, specifically whether subcutaneous dosing enhances drainage to axillary lymph nodes compared to intramuscular injection [62,63], and whether escalating dose priming reproduces improvements seen in macaques [59].

The primary study outcomes include the proportion of participants that develop V3 glycan epitope-specific and BG18 class-specific MBCs, and to determine the BCR immunogenetics (variable heavy allele, variable light allele, and HCDR3 length) and sequences in responders. Leukapheresis will be used for PBMC collection and fine needle aspiration (FNA) will be used to interrogate lymph node B and T follicular helper cells. If successful, this priming regimen will be used in combination with intermediate stage shaping and late-stage wild-type polishing immunogens to further induce promising BG18-like bnAb lineages.

HVTN 309/312 & 307/3XX: testing protein vs. mRNA for two lineage-targeting strategies, CD4bs CH235, and V3 glycan DH270

The HVTN will be testing two lineage-targeting strategies – the CD4bs CH235 bnAb program and the V3 glycan DH270 bnAb program – in both protein nanoparticle and mRNA vaccine platforms. The HVTN 309 study is the first in a series of studies to induce the VH1-46-dependent CD4bs CH235 lineage, with the series testing versions of the CH505 M5.G458K SOSIP prime and CH505 TF chSOSIP boost. The immunogens are based on the UCA for CH235 and CH303, both isolated from the same African individual [64]. HVTN 309 will test a ferritin nanoparticle (FeNP) 24-mer adjuvanted with 3M-052-AF + alum or with the empty lipid nanoparticle (LNP) ACU-026-001-1. HVTN 312 will test mRNA versions of the same immunogens in membrane-bound gp160 versions. Preclinical testing has successfully induced intermediate-step bnAbs in knockin mice and macaques [65▪▪,66] and the mRNA versions may offer some developmental advantages [67].

The second series targets the potent V3 glycan DH270 lineage [68], testing protein NP (HVTN 307) vs. mRNA versions (under development). The prime CH848 10.17DT, a modified transmitted founder virus, features a shortened V1 loop and removal of two V1 glycans (N133 and N138). Using the mutation-guided methodology, boosts are designed to favor induction of key activation-induced cytidine deaminase (AID) cold spot mutations. Preclinical testing induced Tier 2 heterologous neutralization and improbable mutations at HC G75R and LC S27Y in knockin mice [65▪▪], with mRNA providing further LC mutational advantages [69], and boosting contributing further intermediate stage key improbable mutations [38]. The human mRNA study will also include use of a novel chimeric prime with implantation of a V3 sequence from a second V3 bnAb lineage, testing whether two different lineages targeting the V3 glycan can be induced through one hybrid immunogen. A second chimeric priming approach will be tested in HVTN 310 described below.

HVTN 310: an mRNA virus-like particle comprehensive approach for ‘priming-shaping-polishing’

HVTN 310, building on promising results in macaques, will test a series of mRNA-expressed virus-like particle (VLP) immunogens encompassing a ‘priming, shaping, and polishing’ approach within a single study to induce mature VRC01-class and CHO1-class bnAb responses, potentially within the same individual. This approach is based on work by Paolo Lusso et al.[70▪] that showed sequential immunization with mRNA-encoding envelopes derived from three HIV-1 clades (A/B/C), each co-formulated with SIVmac239 Gag mRNA, induced 79% per-exposure risk reduction against multiple intravaginal challenges. A follow-up study in mice was performed with adaptations to improve neutralizing responses [71]. HVTN 310 primes with a VLP expressed Clade C 426c Env missing glycans at positions 276 (loop D), 460, and 463 (V5 loop) around the CD4bs. An alternative prime replaces the 426c V1-V2-V3 loops with these sequences from the Clade A Q23.17 TF virus, designed to engage V2 apex CH01-class germline Abs [72]. Subsequent boosts at months 2, 4, 6, and 8 will use mRNA VLPS with increasingly intact glycans, followed by autologous then heterologous wild-type trimers. The primary outcomes include antigen specific B cells with both VRC01-class and CHO1-class bnAb features, and autologous and heterologous neutralizing responses. This ‘prime to polish’ study will be an important keystone test of the strategy and several important design features important to the program.

ANALYSIS GOALS AND WORKFLOW

B cell assays are the most important immunological interrogation step for the Discovery Medicine program, with induction of paired variable heavy and variable light alleles suggesting epitope specific bnAbs and the development of characteristic mutations providing evidence of bnAb lineage maturation (Fig. 2). Serological studies provide supportive evidence of plasma cell responses, but negative results do not rule out B cell initiation. In addition, interim analyses of adequate, but incomplete data sets are used to make iterative changes.

F2FIGURE 2:

Laboratory analyses. The B cell analysis workflow outlined here includes FACS single cell sorting, BCR sequencing, and mAb cloning. These allow assessment of VH and VL allele usage, development of key mutations, and evaluation of binding affinity (top). Serological assays, which can be run in parallel, provide complementary and more rapid assessments of antibody development, including epitope-specific BAMA, epitope-specific neutralizing assay, and EMPEM (bottom). ++ = cell sorting using two different flourochrome labeled antigens.

B cell analyses

The first important vaccine responses are immunogen-binding and epitope-specific IgG+ B cells. Leukapheresis and lymph node fine needle aspiration are used to collect cells from the periphery and germinal center, respectively, and they are sorted by flow cytometry. Antigen-specific MBCs are then applied to the 10X Genomics single-cell platform and next-generation sequencing (NGS) is used for BCR evaluation [73,74]. Paired variable heavy and variable light sequences are matched for allelic comparison to known antibody classes and CDR3 s sequences are evaluated for germline assignment [23▪▪,75] and key non-activation-induced cytidine deaminase (non-AID) derived mutations important for lineage development [76–78]. Sequencing is the most sensitive measure of vaccine response and is critical for iteration [38,77,78].

Monoclonal antibody (mAb) cloning is then used to measure affinity and determine 3D structural binding characteristics (angle of approach, epitope-paratope interactions). The affinity of mAbs as determined by surface plasmon resonance (SPR) or biolayer interferometry (BLI) [23▪▪]. mAb epitope mapping is then performed using a series of complementary techniques: binding antibody multiplex assay (BAMA), neutralization, and cryo-electron microscopy (for amino acid level mapping) [79,80]. This step links the mAb genetic signature to functional binding, allowing comparisons of sequence, affinity, and angle of approach of model bnAbs such as VRC01, CH235, or BG18. It also allows discovery of new bnAb-like mAbs and is particularly helpful for the epitope-agnostic designs where the outcomes of a polyclonal response are less predictable.

Serum analyses

The serum assays allow a rapid and cost effective assessment of antibody output, including individual response rate and magnitude of polyclonal, epitope-focused, and bnAb class-specific antibodies. The BAMA assesses polyclonal responses and when combined with knockout antigens allows determination of epitope-targeting responses [81]. Pseudo-virus neutralization assays, with and without knockout antigens, are critical for determining functional capacity against the autologous virus [82,83] and assessing development of cross neutralizing breadth [84]. Electron microscope polyclonal epitope mapping (EMPEM) [85], a new technique, adds a wealth of structural information, including epitope specificity and off target responses, angles of approach, and visualization of the dynamics of Ab responses at individual level over time [86▪]. This last feature is useful for prioritizing expensive analyses like sequencing and mAb cloning for best responding individuals.

CONCLUSION

Success for this first round of Discovery Medicine trials is defined by how well they activate bnAb class responses, including whether they induce suggestive variable heavy and variable light alleles and key paratope defining mutations. The next round of studies will test a series of boosting agents to measure how far down the maturation pathway promising lineages can be pushed. To support this, more potent adjuvants are also required to improve the germinal center responses and durability [87,88] and the program has several studies evaluating 3M-052-AF [67,89–91], empty LNP [66,92,93], and SMNP [59,61]. A unified vaccine will likely require combining approaches into a multiepitope inducing prime-boost strategy (Fig. 3). VLPs are one promising option [94], as they provide durable immunogenicity [88], proven efficacy with multiple licenced vacines [95–100], and mRNA versions are attractive due to ease of manufacture, in-vivo expression, and technological advances like dose-sparing self-amplifying RNA [101,102]. Multimeric nanoparticles are another important alternative under investigation for COVID-19 and influenza [103–106] and could be adapted for HIV. Finally, adding a T cell component may be critical for ultimate success [107▪], and trials are already under way testing a promising CD8+ approach [108–110].

F3FIGURE 3:

Hypothetical combination HIV-1 three-epitope vaccine. The table (top) illustrates a three-epitope combination featuring a two-dose prime targeting the CD4bs, V3 glycan, and MPER regions, then two different shaping boosts, followed by a polishing boost, all distributed over 12 months. A final durability boost is hypothesized at 24 months and periodically thereafter to maintain adequate bnAb levels. Two-model platform delivery options include a combination mRNA encoded CD4bs (red), V3 glycan (blue), and MPER (purple) single VLP (bottom left) or multimeric triple epitope nanoparticle (bottom right).

Acknowledgements

The authors would like to thank Lisa Donohue for assistance withFigs. 1–3 and Rachael Parks and Larry Corey for discussions on content and contributions to the manuscript.

Financial support and sponsorship

Funding was provided by NIH grant 3UM1AI068614 to T.M.M. and S.R.

Funding was provided by NIH grant 5UM1AI068635 to Y.H.

Conflicts of interest

T.M.M., S.R., and Y.H. have no conflicts of interest.

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

▪ of special interest

▪▪ of outstanding interest

REFERENCES 1. Haynes BF, Wiehe K, Borrow P, et al. Strategies for HIV-1 vaccines that induce broadly neutralizing antibodies. Nat Rev Immunol 2023; 23:142–158. 2. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 2009; 361:2209–2220. 3. Sok D, Burton DR. Recent progress in broadly neutralizing antibodies to HIV. Nat Immunol 2018; 19:1179–1188. 4. Saphire EO, Parren PW, Pantophlet R, et al. Crystal structure of a neutralizing human IGG against HIV-1: a template for vaccine design. Science 2001; 293:1155–1159. 5. Burton DR, Desrosiers RC, Doms RW, et al. HIV vaccine design and the neutralizing antibody problem. Nat Immunol 2004; 5:233–236. 6. Davenport TM, Gorman J, Joyce MG, et al. Somatic hypermutation-induced changes in the structure and dynamics of HIV-1 broadly neutralizing antibodies. Structure 2016; 24:1346–1357. 7. Haynes BF, Fleming J, St Clair EW, et al. Cardiolipin polyspecific autoreactivity in two broadly neutralizing HIV-1 antibodies. Science 2005; 308:1906–1908. 8. 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. 9. Kwong PD, Mascola JR. Human antibodies that neutralize HIV-1: identification, structures, and B cell ontogenies. Immunity 2012; 37:412–425. 10. Mouquet H, Nussenzweig MC. Polyreactive antibodies in adaptive immune responses to viruses. Cell Mol Life Sci 2012; 69:1435–1445. 11. Wu X, Yang ZY, Li Y, et al. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 2010; 329:856–861. 12. Mascola JR, Haynes BF. HIV-1 neutralizing antibodies: understanding nature's pathways. Immunol Rev 2013; 254:225–244. 13. Zhou T, Georgiev I, Wu X, et al. Structural basis for broad and potent neutralization of HIV-1 by antibody VRC01. Science 2010; 329:811–817. 14. Walker LM, Phogat SK, Chan-Hui PY, et al. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 2009; 326:285–289. 15. Liao HX, Bonsignori M, Alam SM, et al. Vaccine induction of antibodies against a structurally heterogeneous site of immune pressure within HIV-1 envelope protein variable regions 1 and 2. Immunity 2013; 38:176–186. 16. Wu X, Zhou T, Zhu J, et al. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 2011; 333:1593–1602. 17. Kepler TB, Liao HX, Alam SM, 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. 18. Walker LM, Huber M, Doores KJ, et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 2011; 477:466–470. 19. Haynes BG, Burton DR, Mascola JR. Multiple roles for HIV broadly neutralizing antibodies. Sci Transl Med 2019; 11:eaaz2686. 20. Williams WB, Wiehe K, Saunders KO, Haynes BF. Strategies for induction of HIV-1 envelope-reactive broadly neutralizing antibodies. J Int AIDS Soc 2021; 24: (Suppl 7): e25831. 21. Jardine JG, Kulp DW, Havenar-Daughton C, et al. HIV-1 broadly neutralizing antibody precursor B cells revealed by germline-targeting immunogen. Science 2016; 351:1458–1463. 22. Lee JH, Toy L, Kos JT, et al. Vaccine genetics of IGHV1-2 VRC01-class broadly neutralizing antibody precursor naive human B cells. NPJ Vaccines 2021; 6:113. 23▪▪. Leggat DJ, Cohen KW, Willis JR, et al. Vaccination induces HIV broadly neutralizing antibody precursors in humans. Science 2022; 378:eadd6502. 24▪▪. Corey L, Gilbert PB, Juraska M, et al. Two randomized trials of neutralizing antibodies to prevent HIV-1 acquisition. N Engl J Med 2021; 384:1003–1014. 25. Victora GD, Nussenzweig MC. Germinal centers. Annu Rev Immunol 2022; 40:413–442. 26. Abbott RK, Lee JH, Menis S, et al. Precursor frequency and affinity determine B cell competitive fitness in germinal centers, tested with germline-targeting HIV vaccine immunogens. Immunity 2018; 48:133–146. e136. 27. Hagglof T, Cipolla M, Loewe M, et al. Continuous germinal center invasion contributes to the diversity of the immune response. Cell 2023; 186:147–161. e115. 28. Tas JMJ, Koo JH, Lin YC, et al. Antibodies from primary humoral responses modulate the recruitment of naive B cells during secondary responses. Immunity 2022; 55:1856–1871. e1856. 29. Burton DR. Advancing an HIV vaccine; advancing vaccinology. Nat Rev Immunol 2019; 19:77–78. 30. Stamatatos L, Pancera M, McGuire AT. Germline-targeting immunogens. Immunol Rev 2017; 275:203–216. 31. Burton DR. What are the most powerful immunogen design vaccine strategies? Reverse vaccinology 2.0 shows great promise. Cold Spring Harb Perspect Biol 2017; 9:a030262. 32. Jardine J, Julien JP, Menis S, et al. Rational HIV immunogen design to target specific germline B cell receptors. Science 2013; 340:711–716. 33. McGuire AT, Hoot S, Dreyer AM, et al. Engineering HIV envelope protein to activate germline B cell receptors of broadly neutralizing anti-CD4 binding site antibodies. J Exp Med 2013; 210:655–663. 34. Steichen JM, Kulp DW, Tokatlian T, et al. HIV vaccine design to target germline precursors of glycan-dependent broadly neutralizing antibodies. Immunity 2016; 45:483–496. 35. Escolano A, Steichen JM, Dosenovic P, et al. Sequential immunization elicits broadly neutralizing anti-HIV-1 antibodies in Ig Knockin mice. Cell 2016; 166:1445–1458. e1412. 36▪▪. Steichen JM, Lin YC, Havenar-Daughton C, et al. A generalized HIV vaccine design strategy for priming of broadly neutralizing antibody responses. Science 2019; 366:eaax4380. 37. Wiehe K, Bradley T, Meyerhoff RR, et al. Functional relevance of improbable antibody mutations for HIV broadly neutralizing antibody development. Cell Host Microbe 2018; 23:759–765. e756. 38. Wiehe K, Saunders KO, Stalls V, et al. Mutation-guided vaccine design: a strategy for developing boosting immunogens for HIV broadly neutralizing antibody induction. bioRxiv 2022.11.11.516143. 39▪. Luo S, Jing C, Ye AY, et al. Humanized V(D)J-rearranging and TdT-expressing mouse vaccine models with physiological HIV-1 broadly neutralizing antibody precursors. Proc Natl Acad Sci U S A 2023; 120:e2217883120. 40. Sastry M, Changela A, Gorman J, et al. Diverse murine vaccinations reveal distinct antibody classes to target fusion peptide and variation in peptide length to improve HIV neutralization. J Virol 2023; 97:e0160422. 41. Xu K, Acharya P, Kong R, et al. Epitope-based vaccine design yields fusion peptide-directed antibodies that neutralize diverse strains of HIV-1. Nat Med 2018; 24:857–867. 42. Williams WB, Alam SM, Ofek G, et al. Vaccine induction in humans of polyclonal HIV-1 heterologous neutralizing antibodies. medRxiv 2023.03.09.23286943. 43. Li D, Romain G, Flamar AL, et al. Targeting self- and foreign antigens to dendritic cells via DC-ASGPR generates IL-10-producing suppressive CD4+ T cells. J Exp Med 2012; 209:109–121. 44. Kervevan J, Bouteau A, Lanza JS, et al. Targeting human langerin promotes HIV-1 specific humoral immune responses. PLoS Pathog 2021; 17:e1009749. 45. Moodie Z, Rossini AJ, Hudgens MG, et al. Statistical evaluation of HIV vaccines in early clinical trials. Contemp Clin Trials 2006; 27:147–160. 46. Huang Y, Karuna ST, Janes H, et al. Use of placebos in Phase 1 preventive HIV vaccine clinical trials. Vaccine 2015; 33:749–752. 47. Kulp DW, Steichen JM, Pauthner M, et al. Structure-based design of native-like HIV-1 envelope trimers to silence nonneutralizing epitopes and eliminate CD4 binding. Nat Commun 2017; 8:1655. 48. Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2: preliminary report. N Engl J Med 2020; 383:1920–1931. 49. West AP Jr, Diskin R, Nussenzweig MC, Bjorkman PJ. Structural basis for germ-line gene usage of a potent class of antibodies targeting the CD4-binding site of HIV-1 gp120. Proc Natl Acad Sci U S A 2012; 109:E2083–E2090. 50. Zhou T, Zhu J, Wu X, et al. Multidonor analysis reveals structural elements, genetic determinants, and maturation pathway for HIV-1 neutralization by VRC01-class antibodies. Immunity 2013; 39:245–258. 51. Scheid JF, Horwitz JA, Bar-On Y, et al. HIV-1 antibody 3BNC117 suppresses viral rebound in humans during treatment interruption. Nature 2016; 535:556–560. 52. Cohen KW, De Rosa SC, Fulp WJ, et al. A first-in-human germline-targeting HIV nanoparticle vaccine induced broad and publicly targeted helper T cell responses. Sci Transl Med 2023; 15:eadf3309. 53. deCamp AC, Corcoran MM, Fulp WJ, et al. Human immunoglobulin gene allelic variation impacts germline-targeting vaccine priming. medRxiv 2023.03.10.23287126. 54. Medina-Ramirez M, Garces F, Escolano A, 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. 55. Wyatt R, Kwong PD, Desjardins E, et al. The antigenic structure of the HIV gp120 envelope glycoprotein. Nature 1998; 393:705–711. 56. Parks KR, MacCamy AJ, Trichka J, et al. Overcoming steric restrictions of VRC01 HIV-1 neutralizing antibodies through immunization. Cell Rep 2019; 29:3060–3072. e3067. 57. McGuire AT, Gray MD, Dosenovic P, et al. Specifically modified Env immunogens activate B-cell precursors of broadly neutralizing HIV-1 antibodies in transgenic mice. Nat Commun 2016; 7:10618. 58. Cirelli KM, Carnathan DG, Nogal B, et al. Slow delivery immunization enhances HIV neutralizing antibody and germinal center responses via modulation of immunodominance. Cell 2019; 177:1153–1171. e1128. 59. Lee JH, Sutton HJ, Cottrell CA, et al. Long-primed germinal centres with enduring affinity maturation and clonal migration. Nature 2022; 609:998–1004. 60. Tam HH, Melo MB, Kang M, et al. Sustained antigen availability during germinal center initiation enhances antibody responses to vaccination. Proc Natl Acad Sci U S A 2016; 113:E6639–E6648. 61. Silva M, Kato Y, Melo MB, et al. A particulate saponin/TLR agonist vaccine adjuvant alters lymph flow and modulates adaptive immunity. Sci Immunol 2021; 6:eabf1152. 62. Pauthner M, Havenar-Daughton C, Sok D, et al. Elicitation of robust tier 2 neutralizing antibody responses in nonhuman primates by HIV envelope trimer immunization using optimized approaches. Immunity 2017; 46:1073–1088. e1076. 63. Havenar-Daughton C, Carnathan DG, Boopathy AV, et al. Rapid germinal center and antibody responses in nonhuman primates after a single nanoparticle vaccine immunization. Cell Rep 2019; 29:1756–1766. e1758. 64. Gao F, Bonsignori M, Liao HX, et al. Cooperation of B cell lineages in induction of HIV-1-broadly neutralizing antibodies. Cell 2014; 158:481–491. 65▪▪. Saunders KO, Wiehe K, Tian M, et al. Targeted selection of HIV-specific antibody mutations by engineering B cell maturation. Science 2019; 366:eaay7199. 66. Saunders KO, Countis J, Stalls V, et al. Vaccine induction of CD4-mimicking broadly neutralizing antibody precursors in macaques. bioRxiv 2023.03.05.531154. 67. Saunders KO, Verkoczy LK, Jiang C, et al. Vaccine Induction of Heterologous Tier 2 HIV-1 Neutralizing Antibodies in Animal Models. Cell Rep 2017; 21:3681–3690. 68. Bonsignori M, Kreider EF, Fera D, et al. Staged induction of HIV-1 glycan-dependent broadly neutralizing antibodies. Sci Transl Med 2017; 9:eaai7514. 69. Mu Z, Wiehe K, Saunders KO, et al. mRNA-encoded HIV-1 Env trimer ferritin nanoparticles induce monoclonal antibodies that neutralize heterologous HIV-1 isolates in mice. Cell Rep 2022; 38:110514. 70▪. Zhang P, Narayanan E, Liu Q, et al. A multiclade env-gag VLP mRNA vaccine elicits tier-2 HIV-1-neutralizing antibodies and reduces the risk of heterologous SHIV infection in macaques. Nat Med 2021; 27:2234–2245. 71. Zhang P, Falcone S, Tsybovsky Y, et al. Increased neutralization potency and breadth elicited by a SARS-CoV-2 mRNA vaccine forming virus-like particles. Proc Natl Acad Sci U S A 2023; 120:e2305896120. 72. Bonsignori M, Hwang KK, Chen X, et al. Analysis of a clonal lineage of HIV-1 envelope V2/V3 conformational epitope-specific broadly neutralizing antibodies and their inferred unmutated common ancestors. J Virol 2011; 85:9998–10009. 73. Goodwin S, McPherson JD, McCombie WR. Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet 2016; 17:333–351. 74. Zheng GX, Terry JM, Belgrader P, et al. Massively parallel digital transcriptional profiling of single cells. Nat Commun 2017; 8:14049. 75. DeKosky BJ, Kojima T, Rodin A, et al. In-depth determination and analysis of the human paired heavy- and light-chain antibody repertoire. Nat Med 2015; 21:86–91. 76. Goo L, Chohan V, Nduati R, Overbaugh J. Early development of broadly neutralizing antibodies in HIV-1-infected infants. Nat Med 2014; 20:655–658. 77. Lucier A, Fong Y, Li SH, et al. Frequent development of broadly neutralizing antibodies in early life in a large cohort of children with human immunodeficiency virus. J Infect Dis 2022; 225:1731–1740. 78. Muenchhoff M, Adland E, Karimanzira O, et al. Nonprogressing HIV-infected children share fundamental immunological features of nonpathogenic SIV infection. Sci Transl Med 2016; 8:358ra125. 79. Jardine JG, Ota T, Sok D, et al. HIV-1 VACCINES. Priming a broadly neutralizing antibody response to HIV-1 using a germline-targeting immunogen. Science 2015; 349:156–161. 80. Yacoob C, Pancera M, Vigdorovich V, et al. Differences in allelic frequency and CDRH3 region limit the engagement of HIV Env immunogens by putative VRC01 neutralizing antibody precursors. Cell Rep 2016; 17:1560–1570. 81. Yates NL, deCamp AC, Korber BT, et al. HIV-1 envelope glycoproteins from diverse clades differentiate antibody responses and durability among vaccinees. J Virol 2018; 92:e01843-17. 82. Montefiori DC. Measuring HIV neutralization in a luciferase reporter gene assay. Methods Mol Biol 2009; 485:395–405. 83. Todd CA, Greene KM, Yu X, et al. Development and implementation of an international proficiency testing program for a neutralizing antibody assay for HIV-1 in TZM-bl cells. J Immunol Methods 2012; 375:57–67. 84. deCamp A, Hraber P, Bailer RT, et al. Global panel of HIV-1 Env reference strains for standardized assessments of vaccine-elicited neutralizing antibodies. J Virol 2014; 88:2489–2507. 85. Antanasijevic A, Sewall LM, Cottrell CA, et al. Polyclonal antibody responses to HIV Env immunogens resolved using cryoEM. Nat Commun 2021; 12:4817. 86▪. Antanasijevic A, Bowman CA, Kirchdoerfer RN, et al. From structure to sequence: antibody discovery using cryoEM. Sci Adv 2022; 8:eabk2039. 87. McElrath MJ. Adjuvants: tailoring humoral immune responses. Curr Opin HIV AIDS 2017; 12:278–284. 88. Pulendran B, S Arunachalam P, O’Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov 2021; 20:454–475. 89. Fox CB, Orr MT, Van Hoeven N, et al. Adsorption of a synthetic TLR7/8 ligand to aluminum oxyhydroxide for enhanced vaccine adjuvant activity: a formulation approach. J Control Release 2016; 244:98–107. 90. Saunders KO, Lee E, Parks R, et al. Neutralizing antibody vaccine for pandemic and preemergent coronaviruses. Nature 2021; 594:553–559. 91. Smirnov D, Schmidt JJ, Capecchi JT, Wightman PD. Vaccine adjuvant activity of 3M-052: an imidazoquinoline designed for local activity without systemic cytokine induction. Vaccine 2011; 29:5434–5442. 92. Alameh MG, Tombacz I, Bettini E, et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity 2021; 54:2877–2892. e2877. 93. Pardi N, Hogan MJ, Naradikian MS, et al. Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J Exp Med 2018; 215:1571–1588. 94. Lusso P. The quest for an HIV-1 vaccine: will mRNA deliver us from evil? Expert Rev Vaccines 2023; 22:267–269. 95. Guevara A, Cabello R, Woelber L, et al. Antibody persistence and evidence of immune memory at 5years following administration of the 9-valent HPV vaccine. Vaccine 2017; 35:5050–5057. 96. Schiller J, Lowy D. Explanations for the high potency of HPV prophylactic vaccines. Vaccine 2018; 36:4768–4773. 97. Van Damme P, Ward JW, Shouval D, Zanetti A. Plotkin's vaccines. The Netherlands: Elsevier Amsterdam; 2018. 98. Yamaguchi M, Sugahara K, Shiosaki K, et al. Fine structure of hepatitis B virus surface antigen produced by recombinant yeast: comparison with HBsAg of human origin. FEMS Microbiol Lett 1998; 165:363–367. 99. Chu KB, Quan FS. Respiratory viruses and virus-like particle vaccine development: how far have we advanced? Viruses 2023; 15: 100. Lopez P, Lopez-Medina E, Saez-Llorens X, et al. Immunogenicity and tolerability of a bivalent virus-like particle norovirus vaccine candidate in children from 6 months up to 4 years of age: a phase 2 randomized, double-blind trial. Hum Vaccin Immunother 2023; 19:2204787. 101. Schmidt C, Schnierl

留言 (0)

沒有登入
gif