Fact-checked by Grok 2 weeks ago

HIV vaccine development

HIV vaccine development comprises the scientific and clinical pursuits to engineer a preventive against human immunodeficiency virus type 1 (HIV-1), the primary causative agent of acquired immunodeficiency (AIDS). Launched in the early 1980s following HIV-1's isolation, these endeavors have tested myriad designs—including recombinant proteins, viral vectors, and nucleic acid-based platforms—across more than 300 trials, encompassing 11 pivotal efficacy studies, yet have failed to deliver a licensed capable of reliably blocking . Key hurdles stem from HIV-1's extraordinary antigenic variability, driven by error-prone and high replication rates, which generate diverse quasispecies evading conventional antibody and T-cell responses, alongside the virus's capacity to deplete + T cells essential for adaptive immunity. The sole modestly efficacious trial, RV144 in (2009), achieved approximately 31% protection via a canarypox prime and gp120 boost regimen, attributed partly to non-neutralizing antibodies, but follow-up efforts like VAX003 and recent vector-based trials yielded null or negligible results. Promising leads, such as broadly neutralizing antibodies (bNAbs) targeting conserved epitopes and germline-targeting immunogens to initiate bNAb lineages, persist in preclinical and early-phase testing, including mRNA platforms, though phase 3 validations like the 2024 PrEPVacc study confirmed ineffectiveness of tested regimens, highlighting ongoing empirical setbacks and the imperative for novel correlates of protection beyond empirical trial-and-error. Despite four decades of investment yielding insights into immunobiology, the field's stagnation—evident in recent funding reallocations away from certain consortia—underscores causal complexities in inducing sterilizing immunity absent from natural controllers, fueling scrutiny of traditional vaccine paradigms.

Historical Background

Early Research and Initial Efforts (1980s–1990s)

The identification of HIV as the causative agent of AIDS in 1983 by French researchers at the Pasteur Institute, led by Luc Montagnier, and independently in 1984 by Robert Gallo's team at the U.S. National Cancer Institute, spurred immediate vaccine research initiatives. Early efforts emphasized the virus's envelope glycoproteins, particularly gp120 and gp160, as targets for neutralizing antibodies, drawing from successes with other viral vaccines like hepatitis B. In April 1984, U.S. Secretary of Health and Human Services Margaret Heckler publicly stated that an effective AIDS vaccine would be ready for human testing within two years, reflecting initial optimism amid the escalating epidemic but underestimating HIV's mutational rate and integration into host DNA. Preclinical studies in the mid-1980s utilized chimpanzees, the only non-human susceptible to , to test recombinant subunit . These models demonstrated transient against low-dose viral challenges with envelope-based immunogens, but inconsistent results underscored the limitations of extrapolating to humans due to HIV's adaptation to hosts. Vaccine candidates were produced via technology in or mammalian cells to avoid risks of live-virus approaches, given HIV's potential for oncogenicity and persistence. By 1986, informal collaborations, including one organized by Gallo, coordinated global efforts, though challenges like the virus's rapid —exhibiting up to 10% within individuals—were already evident from sequencing data. The first human , a Phase I safety and study, began in August 1987 at the in , involving HIV-uninfected volunteers immunized with a recombinant gp120 protein . This trial, sponsored by the U.S. Army and MicroGenesys, confirmed tolerability but induced only weak, narrow neutralizing responses. Through the , additional Phase I and II trials expanded to candidates like gp160 from Oncogen (Bristol-Myers Squibb) and whole-killed virus preparations, testing adjuvants such as or incomplete Freund's to boost immunity; however, remained modest and strain-specific, with no of protective . By 1997, over a dozen envelope-focused regimens had entered early human testing, primarily in the U.S. and , but preclinical data increasingly revealed HIV's shielding of conserved epitopes, foreshadowing broader setbacks. These efforts, largely driven by pharmaceutical firms like Merck and due to limited initial public funding, established safety profiles but highlighted the need for strategies beyond antibody induction.

Key Milestones in Pre-Clinical and Early Clinical Work

Following the identification of as the causative agent of AIDS in 1984, pre-clinical vaccine research rapidly shifted to animal models, particularly (SIV) in macaques and HIV-1 challenges in chimpanzees, to evaluate candidate immunogens for safety and preliminary efficacy. In 1989, two independent studies reported protection in rhesus macaques: one using formalin-inactivated SIV combined with , which prevented infection upon homologous challenge in four of four animals, and another employing a live-attenuated SIV variant (SIVmac239Δnef), which conferred sterilizing immunity against pathogenic challenge. These results highlighted the potential of both inactivated and attenuated approaches but raised concerns over safety for human use, as attenuated strains carried risks of reversion to . By 1990, pre-clinical work advanced with recombinant envelope glycoprotein gp120, which, when administered to chimpanzees, elicited neutralizing antibodies and protected against low-dose HIV-1 challenge in two of three animals, demonstrating feasibility for subunit vaccines targeting the . In 1992, further studies confirmed durable protection from live-attenuated , with vaccinated animals controlling post-challenge for over a year, though incomplete sterilizing immunity underscored limitations in mimicking human transmission dynamics. These models established correlates like + T-cell responses and antibody-mediated clearance but revealed challenges in translating to HIV strains due to viral diversity. Early clinical trials commenced amid optimism but prioritized safety given HIV's integration into host DNA and lack of natural immunity models. In 1986, French researcher Daniel Zagury conducted the first human trial without regulatory approval, vaccinating himself and six HIV-negative Zairian volunteers with a recombinant expressing HIV-1 gp160 and other proteins; transient immune responses were observed, but ethical lapses and lack of controls limited interpretability. The inaugural regulated Phase 1 trial launched in 1987 at the , testing MicroGeneSys's recombinant gp160 in 81 healthy, HIV-uninfected volunteers; it proved safe with no serious adverse events, inducing modest titers but negligible cellular immunity or neutralization of primary isolates. The NIAID's AIDS Vaccine Evaluation Group (AVEG) initiated its first trial in 1988, evaluating gp120 and gp160 candidates in low-risk volunteers, confirming tolerability while highlighting weak against diverse clades. By 1992, AVEG's inaugural 2 trial targeted high-risk HIV-negative individuals, incorporating risk-reduction counseling alongside vaccines like MN-rgp120; safety was affirmed, but responses waned rapidly, yielding no evidence of in small cohorts. Mid-1990s 1/2 studies explored prime-boost regimens, such as canarypox vectors (ALVAC) expressing antigens followed by gp120 boosts, generating CD4+ and CD8+ T-cell responses in volunteers but failing to elicit broadly neutralizing antibodies against tier-2 viruses. These trials collectively established that early envelope-based candidates induced narrow, strain-specific immunity insufficient for broad efficacy, prompting shifts toward T-cell-focused designs while exposing gaps in correlates of .

Biological and Immunological Challenges

HIV's Evasion Mechanisms and Genetic Variability

HIV-1's enzyme operates without proofreading capability, yielding a of approximately 3 × 10^{-5} substitutions per per replication cycle, which is substantially higher than that observed in most other viruses. This elevated error rate, compounded by frequent recombination events during co-infection with multiple viral strains, generates extensive within a single host. Consequently, HIV-1 populations form dynamic quasispecies—clouds of closely related variants differing by multiple mutations—allowing the virus to maintain a of potential escape forms that can be selected under immune pressure. The envelope glycoprotein (), particularly the gp120 subunit, exemplifies this variability, with hypervariable regions (–V5 loops) exhibiting mutation rates up to 1–2% per year in chronic infection, far exceeding conserved genomic regions. These drive antigenic drift, enabling the virus to evade neutralizing antibodies by altering key epitopes or by accumulating N-linked glycans that shield vulnerable sites on the Env trimer. For cellular immunity, genetic changes in epitopes presented by molecules allow escape from cytotoxic T lymphocytes (CTLs), as variant peptides fail to bind HLA alleles or trigger recognition by existing T-cell receptors. Such adaptations occur rapidly, often within weeks of initial immune targeting, perpetuating chronic infection. Beyond raw variability, HIV-1 leverages mutations in accessory genes to counteract intrinsic host defenses, enhancing overall evasion. For instance, polymorphisms in vif inhibit APOBEC3G-mediated hypermutation, while nef variants downregulate MHC-I expression on infected cells, reducing CTL visibility without triggering compensatory NK cell activation. Similarly, vpu mutations counteract tetherin to facilitate virion release. This genetic plasticity positions HIV-1 near its error threshold, balancing diversity generation with functional integrity to sustain replication amid host pressures. The resultant intra- and inter-clade diversity—spanning over 30% nucleotide divergence in env across global subtypes—poses profound barriers to vaccine-induced immunity capable of broad coverage.

Limitations of Animal Models and Correlates of Protection

Animal models for vaccine development face significant limitations due to the virus's human-specific tropism and the inability of most species to naturally replicate and . Chimpanzees, the only non-human susceptible to , rarely progress to AIDS-like , limiting their utility for studying and vaccine efficacy; ethical concerns have further restricted their use since the early 2000s. models employing () or chimeric SHIV viruses better mimic AIDS progression but diverge in immune responses, mucosal transmission routes, and viral genetics from human , leading to poor translational predictive value. Humanized mouse models, such as or hu-HSC mice engrafted with human hematopoietic cells, enable HIV-1 replication but suffer from incomplete immune reconstitution, lack of lymphoid architecture, and variable engraftment efficiency, precluding reliable assessment of -induced protection against systemic or mucosal challenges. These models often overestimate or underestimate immunogenicity due to absent human-specific factors like mucosal immunity and T-cell trafficking, contributing to discrepancies between preclinical success and clinical failures, as seen in the STEP trial where an adenovirus-5 vectored protected macaques but increased HIV acquisition risk in humans. The identification of correlates of protection (CoP)—immune parameters predictive of vaccine efficacy—remains elusive for -1, complicating preclinical evaluation in animal models. Unlike vaccines for pathogens with sterilizing immunity (e.g., neutralizing antibodies for ), HIV lacks defined natural protective immunity, with no single marker like T-cell counts or broadly neutralizing antibodies (bnAbs) consistently correlating with prevention of infection across trials. In the RV144 trial, the only partially efficacious vaccine (31% efficacy in 2009), modest included low Env-specific IgA and high ADCC-mediating IgG3 antibodies, but these have not replicated in subsequent studies, highlighting context-dependency and the challenge of inducing durable, multi-epitope responses against HIV's hypervariability. Animal models exacerbate this by failing to capture human-specific , such as bnAb maturation requiring prolonged exposure absent in short-term challenges, resulting in vaccines that elicit promising responses in non-human primates but fail to protect humans. Overall, these limitations underscore the need for human-centric approaches, as animal-derived have historically misled development efforts despite decades of investment.

Vaccine Design Strategies

Traditional Approaches (e.g., Whole-Virus, Subunit Vaccines)

Subunit vaccines targeting envelope glycoproteins, such as recombinant gp120 or gp160, formed the cornerstone of early traditional efforts, inspired by the success of protein-based vaccines like . These candidates aimed to elicit neutralizing antibodies against the viral , but clinical outcomes revealed limitations in addressing HIV's conformational epitopes and sequence diversity. The first phase I trial of a gp160 subunit vaccine occurred in 1987, yet subsequent evaluations showed no significant efficacy in preventing . The AIDSVAX vaccine, a monomeric gp120 protein formulation developed by VaxGen, advanced to phase III testing in two trials: Vax003 (conducted 2000–2003 in with over 16,000 participants at risk) and Vax004 (1998–2002 in and with about 5,400 participants). Both trials failed to demonstrate protective efficacy, with infection rates similar between vaccinated and placebo groups—yielding adjusted efficacy estimates of -3% to 0% after unblinding and intent-to-treat analysis. Post-trial analyses attributed the lack of success to the vaccine's induction of non-neutralizing antibodies focused on variable loops, which failed to block diverse HIV strains or mucosal transmission. Whole-virus approaches, including inactivated preparations, were pursued cautiously due to HIV's into genomes and potential for incomplete inactivation leading to latent reservoirs or oncogenicity. Unlike successful inactivated for pathogens like , HIV's enveloped structure complicates antigen preservation during beta-propiolactone or formalin treatment, often resulting in diminished . A phase I trial of a genetically modified, inactivated whole-HIV-1 began in 2016, confirming safety in low doses but eliciting only modest responses insufficient for broad protection; further advancement stalled amid concerns over scalability and regulatory hurdles for handling replication-competent residuals. Live-attenuated whole-virus strategies, effective in SIV-macaque models via nef deletion, were deemed too risky for humans given documented cases of disease progression in exposed infants. These traditional methods underscored HIV's resistance to antibody-centric responses alone, as subunit vaccines generated high-titer binding antibodies without the breadth needed against glycan-shielded conserved sites, while whole-virus formats struggled with safety-immunogenicity trade-offs. By the early , failures prompted shifts toward combination regimens incorporating T-cell priming, though pure traditional designs yielded no licensed products.

Novel Approaches Targeting Broadly Neutralizing Antibodies (bnAbs)

Broadly neutralizing antibodies (bnAbs) target conserved epitopes on the HIV-1 envelope glycoprotein (), offering potential for vaccines that neutralize diverse viral strains. Unlike conventional antibodies, bnAbs require extensive and specific maturation pathways, which natural infection rarely elicits efficiently. Novel vaccine strategies focus on engineering immunogens to initiate and guide the development of bnAb precursor B cells, addressing HIV's evasion tactics such as hypervariability and shielding. Germline-targeting approaches design immunogens to bind receptors resembling unmutated bnAb ancestors, priming rare precursor lineages for subsequent boosting. For instance, eOD-GT8 immunogens specifically activate VRC01-class bnAb precursors by mimicking the . In nonhuman studies, germline-targeting induced neutralizing antibodies in germline-reverted models, demonstrating activation of bnAb lineages. Human phase I trials, such as those evaluating BG505 SOSIP trimers modified for germline , have initiated to test and , with early data showing precursor responses but limited breadth. These strategies acknowledge the low frequency of bnAb precursors in humans, estimated at 1 in 10^5 to 10^6 , necessitating precise presentation. Sequential immunization protocols administer a series of tailored immunogens to iteratively mature bnAb responses, mimicking the prolonged antigen exposure in elite controllers. Preclinical models using nanoparticle-displayed trimers or stabilized SOSIP variants have elicited tier-2 neutralizing activity against autologous strains in rabbits and after multiple boosts. A 2022 human trial demonstrated vaccine-induced heterologous neutralizing precursors, marking proof-of-concept for rapid lineage activation. However, achieving the 30-50% mutations typical of potent bnAbs remains challenging, with probabilities of full maturation low without optimized sequencing. Structure-based design leverages cryo-electron microscopy and to engineer native-like Env trimers that expose bnAb while occluding non-neutralizing sites. SOSIP-stabilized trimers, such as BG505 SOSIP.664, adopt prefusion conformations and have been iteratively refined to enhance precursor affinity. Recent advances include cyclically permuted gp120 trimers and computationally optimized variants that select for functional bnAb mutations during affinity maturation. In 2024, engineered activated diverse bnAb precursors in animal models, improving -specific responses. Despite progress, clinical translation faces hurdles, including immunogen stability, delivery vectors like mRNA or adenoviral platforms, and the need for multi- targeting to cover HIV's diversity. Ongoing trials emphasize combinatorial regimens to broaden coverage beyond single bnAb classes.

Emerging Technologies (e.g., mRNA and Vector-Based Platforms)

mRNA platforms enable rapid encoding of HIV envelope () immunogens to stimulate B cells, particularly through germline-targeting strategies that activate rare precursor B cells for broadly neutralizing antibodies (bnAbs). In May 2025, data from two phase 1 trials (IAVI G001 and Fred Hutchinson's HVTN 133) demonstrated proof-of-concept for this approach, showing successful priming of VRC01-class bnAb precursors in humans using sequential mRNA immunogens like eOD-GT8 and Core-g28v2. These trials involved 54 and 18 participants, respectively, with vaccines eliciting immune responses without severe adverse events, marking the first human evidence of targeted bnAb lineage activation. Further advancements include membrane-anchored constructs delivered via mRNA, tested in preclinical models and early human studies as of July 2025, which enhanced B-cell responses by stabilizing trimeric structures to mimic native virus. An 2025 phase 1 in , sponsored by IAVI, began evaluating mRNA-1644 (eOD-GT8 60mer) and related boosters in 120 adults, including people living with , to assess safety and for bnAb induction. Germline-targeting mRNA strategies address 's shielding of conserved epitopes by iteratively boosting immature B cells toward mature bnAbs, with 2025 research confirming simultaneous priming of multiple bnAb classes in animal models. Viral vector platforms, such as and vesicular stomatitis virus (VSV), deliver genes to induce T-cell and antibody responses, leveraging self-adjuvanting properties for durable immunity. Despite historical risks with adenovirus 5 (Ad5) vectors—linked to increased acquisition in the 2007 STEP trial due to + T-cell enhancement—newer serotypes like Ad26 and gorilla-derived mitigate pre-existing immunity and safety concerns. In January 2024, ReiThera, the Ragon Institute, and IAVI initiated preclinical work on a vector encoding highly networked T-cell epitopes, advancing toward phase 1 for broad control via cytotoxic T lymphocytes. IAVI's VSV vector candidates target T-cell epitopes alongside Env immunogens, showing immunogenicity in phase 1 trials post-2020 without the integration risks of DNA platforms. These vectors support multi-epitope designs for conserved HIV regions, with plug-and-play adaptability allowing rapid iteration against variants, as evidenced in 2025 reviews of recombinant viral chassis. Combined mRNA-vector regimens are under exploration to synergize antibody priming with T-cell augmentation, though challenges persist in scaling bnAb maturation and avoiding vector-induced enhancement.

Clinical Trials and Outcomes

Phase I and II Trials: Safety and Immunogenicity

Phase I and II clinical trials of vaccine candidates have evaluated in small cohorts of HIV-uninfected volunteers, followed by assessments in expanded groups to detect and T-cell responses without measuring . These trials, numbering over 100 since the late 1980s, have consistently shown that most candidates—ranging from subunit proteins to viral vectors—are well-tolerated, with adverse events limited to mild-to-moderate injection-site reactions, fatigue, and transient fever in 10-50% of participants, and no vaccine-attributable serious events or infections directly linked to . The inaugural Phase I trial, launched in 1987 by the National Institutes of Health, tested a recombinant gp160 envelope subunit vaccine in 138 healthy volunteers, confirming safety through absence of serious adverse effects and inducing detectable HIV-specific antibodies, though responses were narrow and immunogen-specific without broad neutralization. Early 1990s Phase I/II trials of gp120-based vaccines, such as AIDSVAX B/E, administered at doses up to 600 μg, similarly demonstrated tolerability with primarily local reactogenicity, eliciting peak anti-gp120 binding antibody titers of 1:10,000-1:50,000 in 70-90% of recipients after three doses, but these waned within 6-12 months and targeted only homologous strains. From the mid-1990s onward, prime-boost strategies—often DNA priming followed by (e.g., canarypox ALVAC) or protein boosting—dominated Phase I/II evaluations, enhancing cellular over standalone approaches; for instance, ALVAC-HIV (vCP1521) plus gp120 trials in the late 1990s to 2000s induced IFN-γ-secreting T cells in 30-60% of participants and IgG responses, with regimens showing no excess safety signals beyond grade 1-2 events like in 20-40% of cases. Adenoviral vector trials, such as those with Ad26 mosaics in the , reported comparable safety profiles and broader + T-cell responses targeting conserved epitopes in up to 80% of vaccinees, though induction remained focused on non-neutralizing epitopes. Despite these advances, across trials has proven insufficient for sterilizing immunity, with humoral responses rarely exceeding 10-20% neutralization breadth against diverse clades and cellular responses declining to baseline within 1-2 years, underscoring 's evasion via shielding and hypervariability. Recent I/II efforts post-2010, including stabilized Env trimers and mRNA platforms, continue to prioritize while targeting B-cell activation for bnAbs, yielding higher-tier neutralization in subsets (e.g., 20-40% of participants) but still transient and low-magnitude compared to natural controllers. This pattern of safe yet suboptimal immune elicitation has informed progression to larger trials, revealing no evidence of enhanced susceptibility in recipients.

Phase III Efficacy Trials: Successes and Failures

The first Phase III efficacy trials for vaccines, conducted in the late 1990s and early 2000s, tested recombinant gp120 subunit vaccines but demonstrated no protective . Vax004, initiated in 1998 and involving 5,409 participants at higher risk in and , used bivalent gp120 (B and E subtypes) with MF59 ; it failed to reduce HIV acquisition, with infection rates of 3.8% in vaccinees versus 3.3% in recipients over 36 months. Similarly, Vax003, conducted from 1999 to 2003 in with 2,454 injecting drug users, employed gp120 (B/E) with and also showed no , as HIV incidence was comparable between groups (6.7 infections per 100 person-years in both). These failures highlighted the insufficiency of gp120-based approaches in eliciting broadly protective immune responses against diverse HIV strains. A landmark partial success emerged from the RV144 , conducted in from 2003 to 2009 with 16,402 low-risk adults. This prime-boost regimen combined ALVAC-HIV (vCP1521, a canarypox expressing gp120, gag, and from clades B and E) priming with AIDSVAX B/E (gp120 boost) and showed 31.2% (95% CI: 1.1-51.2; p=0.04) against acquisition at 42 months, with efficacy peaking at 60% in the first year before waning. No effect on or CD4 counts was observed in breakthrough infections, and correlates analysis later linked reduced risk to IgG antibodies against V1V2 scaffold and low Env-specific IgA levels, suggesting a role for non-neutralizing antibodies in mucosal protection. Despite its modest and transient protection, RV144 remains the only Phase III to date demonstrating statistically significant , renewing interest in poxvirus- strategies but underscoring the need for durable responses. Subsequent trials targeting T-cell responses largely failed, often revealing unintended risks. The STEP (HVTN 502) trial, launched in 2004 with 3,003 HIV-uninfected men who have sex with men or at risk in and , tested a Merck Ad5-gag/pol/nef trivalent vaccine; it was halted in 2007 after interim analysis showed no efficacy (hazard ratio [HR] 1.0 overall) and an increased infection risk (HR 2.3) among Ad5-seropositive, uncircumcised participants, attributed to vector-induced enhancement of HIV susceptibility via CD4+ T-cell targets. The parallel Phambili (HVTN 503) trial in , using the same regimen in 801 participants, was stopped early in 2007 with no efficacy observed (HR 1.81 in early unblinded data), reinforcing Ad5 vector limitations due to pre-existing immunity. HVTN 505, a 2010-2013 trial in 2,294 men, employed DNA priming followed by Ad5 boosting (with env, gag, pol inserts); it failed to prevent infection or control viremia (HR 1.21) and was terminated early, further evidencing challenges in T-cell-focused designs against HIV's mutational .
TrialStart YearLocation/ParticipantsRegimenEfficacy OutcomeKey Issues
Vax0041998/ (5,409 high-risk)gp120 (B/E) + MF590% (no reduction)Narrow subtype focus; no broad neutralization
Vax0031999 (2,454 IDUs)gp120 (B/E) + 0% (no reduction)Inadequate immune breadth in high-risk group
RV1442003 (16,402 low-risk)ALVAC prime + gp120 boost31.2% (waning)Modest success; V1V2 IgG correlate identified
STEP (HVTN 502)2004// (3,003 MSM/at-risk)Ad5 gag//nef0%; HR 2.3 in subgroupVector immunity-enhanced risk
Phambili (HVTN 503)2005 (801)Ad5 gag//nef0% (HR 1.81)Pre-existing Ad5 antibodies; early halt
HVTN 5052010USA (2,294 MSM)DNA prime + Ad5 boost0% (HR 1.21)No impact on acquisition or control
Later Phase III efforts adapting RV144 for clade C strains, such as HVTN 702 (Uhambo) from 2016-2020 in with 5,414 women, replicated the ALVAC-AIDSVAX regimen but yielded no (HR 0.97) and was discontinued, possibly due to clade mismatches, population differences, or insufficient in southern cohorts. These repeated failures across six major trials—contrasted with RV144's —underscore HIV's formidable barriers, including antigenic and immune evasion, prompting shifts toward antibody-focused designs while revealing biases in trial assumptions favoring T-cell over humoral responses without durable correlates. No Phase III has yet achieved >50% or demonstrated prevention in diverse global populations.

Recent and Ongoing Trials (Post-2020 Developments)

The phase 2b Imbokodo trial (HVTN 705), evaluating a Janssen-developed adenovirus vector vaccine (Ad26.Mos4.HIV) boosted with clade B/C envelope protein, was halted in January 2022 after an interim analysis showed no significant reduction in HIV infections among 2,624 South African women, with only 4% efficacy observed against the anticipated 50-75%. Similarly, the phase 3 Mosaico trial (HVTN 706), testing an updated version of the same regimen with multi-clade coverage in diverse populations across the Americas and Europe, was discontinued in December 2023 following futility analysis, as it failed to demonstrate protection against HIV acquisition in over 10,000 participants. The phase 2b PrEPVacc trial, assessing two regimens involving DNA prime, modified vaccinia Ankara (MVA-B), and gp120 boosts in 2,114 women in South Africa and Uganda, concluded vaccinations in November 2023 and reported in July 2024 no overall reduction in HIV incidence, though subgroup analyses suggested modest clade C-specific effects insufficient for broad efficacy. These failures underscored persistent challenges in eliciting protective immunity against diverse HIV strains, prompting a pivot from traditional vector-based approaches to strategies targeting broadly neutralizing antibodies (bnAbs). Post-2020 efforts have emphasized early-phase trials for bnAb induction via targeting, which aims to activate rare naive B cells capable of maturing into bnAb producers. In May 2025, combined data from IAVI's phase 1 G002 and G003 trials demonstrated proof-of-concept for this approach, showing vaccination with engineered immunogens successfully primed VRC01-class bnAb precursor B cells in healthy adults without adverse events dominating the response. Building on this, IAVI initiated a phase 1 trial in August 2025 in enrolling approximately 120 adults (including those living with ) to evaluate safety and of a novel candidate across HIV statuses. Parallel advances in mRNA platforms include July 2025 preclinical-to-clinical transitions where membrane-anchored envelope immunogens elicited targeted neutralizing responses in phase 1 settings, bypassing some maturation hurdles. These trials, while promising for , remain in exploratory stages with no data, reflecting a cautious rebuilding after large-scale disappointments. As of 2025, over 20 vaccine candidates are in clinical testing, predominantly phase 1 and 2 focused on bnAb lineages, mRNA-encoded antigens, and sequential boosting regimens, with projected completions for select mRNA trials by 2027. No phase 3 studies are actively recruiting, highlighting a research crossroads where foundational immune correlates must be validated before scaling.

Economic and Resource Allocation Issues

The primary funding sources for HIV vaccine development have historically been dominated by public sector investments, particularly from the U.S. (NIH) through its National Institute of Allergy and Infectious Diseases (NIAID), which accounted for approximately 70% of the global total in 2022, or about $518 million out of $740 million spent worldwide on preventive HIV vaccine research and development (R&D). Philanthropic organizations, led by the Bill & Melinda Gates Foundation, have provided significant support, including $287 million across 16 grants to establish the Collaboration for AIDS Vaccine Discovery (CAVD) in the mid-2000s and a $55 million investment in a 2023 collaboration for HIV vaccine candidates. Other contributors include international bodies such as the Global HIV Vaccine Enterprise and entities like IAVI, which receive diversified funding from governments, philanthropies, and academic partners to advance preclinical and clinical efforts. Private sector involvement from pharmaceutical companies remains limited, often confined to partnerships due to the high-risk profile and lack of commercial success to date. Global investment trends in vaccine R&D have shown stagnation and recent declines amid repeated clinical setbacks and shifting priorities toward alternative prevention modalities like long-acting . In 2021, worldwide spending on preventive vaccines reached $795 million, representing the largest share of the $1.25 billion total for prevention R&D, though this marked a $30 million drop from the prior year. By 2022, the figure fell to approximately $731–740 million, with the U.S. funding nearly 90% of the global total, underscoring heavy reliance on NIH support. A sharp downturn emerged in 2025, as NIAID announced the non-renewal of funding for key consortia, including the Consortia for HIV/AIDS Vaccine Development (CHAVD), which had received an annual $67 million allocation representing a substantial portion of NIH's vaccine efforts; this decision initiated a one-year wind-down period, prompting researchers to seek private alternatives amid an "uncertain future" for the field. European investments bucked the trend modestly, rising 47% to $27.5 million in recent years, but overall philanthropic contributions have waned as foundations redirect toward implementation of proven tools like lenacapavir injectables. These cuts coincide with broader NIH budget reallocations favoring "implementation science" for existing preventives over foundational vaccine research, potentially exacerbating the field's challenges given the absence of an efficacious vaccine after over $10 billion invested globally since the 1980s.

Cost-Benefit Analysis and Prioritization Debates

Over four decades of research, vaccine development has consumed substantial resources, with global investments in HIV prevention R&D, including vaccines, totaling approximately $20 billion from 2000 to 2020, a significant portion directed toward vaccine efforts despite repeated trial failures. In 2022, the alone allocated nearly $600 million to vaccine research, representing 81% of worldwide expenditure on the topic, though funding has shown signs of stagnation or decline, exemplified by the National Institutes of Health's decision in May 2025 to eliminate the $67 million annual budget for the Consortia for Vaccine Development program, which supported collaborative research across multiple institutions. These expenditures reflect high research and development costs driven by the virus's , the need for large-scale clinical trials in high-incidence populations, and challenges in inducing durable immunity, contrasting with more straightforward vaccine successes like those for SARS-CoV-2. Economic models project that a successful vaccine could yield favorable cost-effectiveness ratios under optimistic scenarios of moderate efficacy and scalability. For instance, dynamic transmission models estimate an (ICER) of $42,473 per (QALY) gained for a in high-risk settings like , , deemed cost-effective against a $150,000/QALY , with potential offsets from reduced antiretroviral therapy (ART) and (PrEP) expenditures totaling hundreds of millions in averted healthcare costs. Another analysis posits that a priced at $15 per dose could achieve an ICER of $13,746 per QALY in generalized epidemic contexts, with broader societal benefits including medical savings exceeding $25 per person across over 700 million individuals for a 75% efficacious formulation. However, these projections hinge on unproven assumptions of performance and delivery, as no has yet demonstrated sustained efficacy in phase III trials, raising questions about the reliability of such forward-looking estimates given historical overoptimism in preclinical data. Prioritization debates center on opportunity costs, weighing HIV vaccine pursuits against alternative prevention modalities and other global health threats with higher feasibility of breakthroughs. Proponents argue for continued investment due to HIV's persistent burden—1.3 million new infections annually—and the limitations of , such as adherence challenges and incomplete coverage, with models suggesting a vaccine could avert transmissions more comprehensively than current tools, even amid advances like twice-yearly lenacapavir injections available at low cost ($40/year) in low- and middle-income countries from 2027. Critics, however, highlight the field's low yield after billions invested, advocating reallocation to scalable interventions like expanded and , which have driven incidence declines in prioritized regions; for example, a mere 3% annual drop in coverage could precipitate 8,618 excess U.S. infections and $3.6 billion in added lifetime costs by modeling reduced prevention efficacy. This tension is amplified by recent U.S. funding shifts, including 2025 cuts to preventive programs, signaling a pragmatic reassessment that favors immediate-impact strategies over high-risk, long-horizon vaccine research, particularly when spillover benefits from HIV studies (e.g., mRNA platforms) have arguably been overstated relative to direct outputs. Further contention arises in comparing HIV vaccine prioritization to other infectious diseases, where vaccines like those for or succeeded rapidly due to less mutational evasion, underscoring HIV's unique biological barriers—such as rapid evolution and latency—that inflate risks without commensurate guarantees. While HIV's socioeconomic toll justifies targeted funding, empirical evidence of stalled progress prompts calls for diversified portfolios, with some analyses indicating that resources diverted to more tractable pathogens or HIV cures (e.g., gene editing) might yield higher returns on investment, especially as has transformed HIV into a manageable for many. These debates underscore a core tension: the ethical imperative to pursue eradication tools versus fiscal realism in allocating scarce funds amid competing unmet needs, with recent policy actions like the 2025 NIH cuts reflecting a data-driven pivot toward proven preventives over speculative platforms.

Ethical and Controversial Aspects

Trial Design Ethics in High-Risk Populations

High-risk populations, such as men who have sex with men (MSM), female sex workers, and people who inject drugs, are preferentially recruited for HIV vaccine efficacy trials due to their elevated incidence rates, which enable statistically powered studies with fewer participants and shorter durations compared to low-risk groups. This approach aligns with scientific necessity but raises ethical concerns under principles of and non-maleficence, as these groups often face intersecting vulnerabilities including , legal marginalization, and limited access to healthcare. Informed consent processes in these trials must ensure , yet challenges persist, particularly in ensuring comprehension and voluntariness among participants with lower , economic pressures, or cultural barriers. For instance, studies in MSM and sex worker cohorts have documented incomplete understanding of trial risks, such as potential immune enhancement or social harms like , despite structured consent procedures. Ethical guidelines mandate tailored , independent advocates, and ongoing assessments to mitigate , with trial designs incorporating advisory boards to address power imbalances. The use of placebo controls in high-risk populations has been contentious, especially post-2010s with availability; placebos are deemed unethical if withholding proven interventions like increases infection risk without scientific justification. Revised UNAIDS/WHO guidance emphasizes providing all state-of-the-art prevention (e.g., , condoms) to both arms, treating placebo use as acceptable only when no effective alternative exists or for establishing causality in novel contexts, while monitoring for unintended harms like behavioral disinhibition. In practice, many phase IIb/III trials now integrate to uphold beneficence, though this complicates efficacy endpoints by reducing incident infections. Broader ethical scrutiny includes equitable benefits post-trial, such as access to vaccines if successful, and avoiding in low- and middle-income countries where high-risk groups bear disproportionate trial burdens. Sponsors must monitor social risks, including from perceived trial participation signaling high-risk behavior, and implement post-trial care to prevent long-term vulnerabilities. These considerations, informed by historical trial setbacks like unmitigated infections in early studies, underscore the need for rigorous oversight to balance scientific imperatives with participant protection.

Debates on Risk-Benefit and Alternative Prevention Methods

The risk-benefit profile of vaccine candidates has been intensely debated due to instances of vaccine-enhanced disease in clinical trials. The STEP trial (2004–2007), evaluating a Merck adenovirus 5 (Ad5)-vectored targeting Gag, , and Nef proteins, not only failed to demonstrate efficacy but also showed increased acquisition rates among certain subgroups, particularly uncircumcised men with pre-existing Ad5 neutralizing antibodies. Extended follow-up confirmed a of approximately 2.0 for infection in this cohort during the first 18 months post-vaccination, attributed to potential depletion of + T cells or altered mucosal immunity that facilitated viral entry. These findings halted the trial early and prompted scrutiny over whether vector-specific immune responses could inadvertently heighten susceptibility, raising ethical questions about proceeding with similar platforms without resolving enhancement mechanisms. Proponents of continued vaccine investment emphasize the theoretical long-term benefits of inducing broadly neutralizing antibodies (bnAbs) or cellular immunity for sterilizing protection, potentially obviating daily interventions and addressing non-adherence rates exceeding 50% in real-world settings. However, skeptics argue that repeated Phase III failures—such as the 31% efficacy in the 2009 RV144 trial that waned rapidly—and the absence of a licensed after over four decades justify reallocating resources, given the empirical success of alternatives like (PrEP). with daily oral tenofovir disoproxil fumarate-emtricitabine reduces acquisition risk by 99% under optimal adherence, while newer long-acting options like (every two months) and (every six months, FDA-approved June 2025) achieve 89–96% efficacy in men and gender-diverse populations, mitigating adherence barriers through subcutaneous injections. Debates intensify over prioritization in high-incidence regions, where scale-up has stalled due to costs (e.g., $100–300 annually per user in low-income countries) and issues, yet still averts more infections than speculative in modeling studies. Complementary methods, including voluntary medical male (50–60% risk reduction for heterosexual men in randomized trials) and antiretroviral treatment as prevention (undetectable equates to untransmissibility per the studies), offer population-level impact without the uncertainties of vaccine-induced enhancement or variable across clades. Critics of vaccine-centric funding, which exceeded $1 billion annually pre-2020 from sources like the U.S. NIH, contend it diverts from behavioral interventions and diagnostics, though advocates counter that could achieve thresholds unattainable by individual-level tools amid 1.3 million new infections in 2024. Empirical data thus frames the tension: proven preventives like excel in adherent users but falter globally, while promise universality yet carry unresolved risks substantiated by trial harms.

Future Directions and Realistic Prospects

Advances in bnAb Induction and Germline Targeting

Germline targeting strategies seek to overcome the rarity and low affinity of receptors for broadly neutralizing antibody (bnAb) precursors by engineering (Env) immunogens that specifically bind unmutated versions of these receptors, thereby priming the B cells for subsequent maturation through boosting immunogens. This multi-stage approach mimics the prolonged affinity maturation observed in natural infection but accelerates it via sequential vaccinations, addressing the high (often >30%) required for bnAbs to penetrate 's glycan shield and neutralize diverse strains. Initial efforts targeted VRC01-class bnAbs directed at the CD4-binding site, with immunogens like the engineered outer (eOD-GT8) demonstrating activation of precursors in animal models as early as 2016, paving the way for human translation. Preclinical advances have expanded to multiple bnAb classes, including V3-glycan and MPER-directed antibodies. In 2024, MPER -liposome immunogens induced polyclonal lineages resembling mature bnAbs and their precursors in nonhuman , marking a shift toward lipid-anchored antigens for enhanced recruitment. Concurrently, germline-targeting Env SOSIP trimers, such as the 2025-designed 3nv.2 variant, were characterized to simultaneously present CD4-binding site, V3, and epitopes, eliciting responses against conserved sites in mice and rabbits without off-target dominance. These designs incorporate stabilizing mutations and glycan modifications to expose vulnerable epitopes while minimizing immunodominant decoys, validated through and sorting assays. Human clinical data provide proof-of-concept for bnAb precursor induction. Two phase 1 trials reported in May 2025 demonstrated that germline-targeting vaccines, including eOD-GT8 and core-gT variants, successfully primed in healthy adults, with detected expansions in 20-97% of participants depending on the , as measured by antigen-specific B cell sequencing and neutralization assays. A separate 2025 study extended this to simultaneous priming of multiple bnAb classes (e.g., CD4-binding and V2-apex) in humans using tailored trimers, though autologous neutralization remained limited without boosting. These trials, conducted by consortia like IAVI and , enrolled low-risk volunteers and confirmed safety, with mild reactogenicity akin to other Env vaccines. Challenges persist in scaling to broad neutralization, as primed precursors require optimized boosting regimens to achieve the 50-70% mutation levels of potent bnAbs, and rare precursor frequencies (1 in 10^5-10^6 s) demand high immunogen doses or adjuvants like 3M-052. Ongoing efforts integrate mRNA platforms for rapid ; a July 2025 trial using mRNA-encoded membrane-anchored Env elicited precursor responses but highlighted the need for multi-dose schedules to drive maturation. Despite these hurdles, targeting has validated the feasibility of directing human repertoires toward bnAb pathways, informing next-generation trials combining priming with lineage-specific boosters by 2026.

Integration with Other HIV Prevention Tools

An effective HIV vaccine, once developed, is anticipated to complement existing biomedical, behavioral, and structural prevention modalities rather than replace them, forming part of a layered or combination strategy to maximize population-level protection against acquisition. Current tools such as (PrEP) with oral tenofovir disoproxil fumarate/emtricitabine (demonstrating 99% efficacy in adherent men who have sex with men and heterosexual couples), long-acting injections (96% efficacy in women and 89% in men who have sex with men and individuals in phase 3 trials), male medical (reducing heterosexual acquisition by 60% in randomized trials), and antiretroviral therapy (ART) for treatment as prevention (TasP, suppressing viral load to undetectable levels and virtually eliminating transmission per the and studies) address immediate risks but face limitations like adherence requirements and incomplete coverage of transmission routes. Mathematical modeling indicates that even a modestly efficacious (e.g., 30-50% effective, as seen in the RV144 trial's 31.2% against HIV-1 acquisition in participants from 2003-2009) could synergize with these tools, potentially averting 20-50% more infections over a decade when combined with scaled-up diagnosis, , and compared to non-vaccine scenarios, particularly in generalized epidemics. Integration would mitigate 's adherence challenges—where real-world drops to 44-74% due to inconsistent use—by offering durable, potentially sterilizing or risk-reducing immunity without daily intervention, while could bridge gaps during vaccine waning or in non-responders. Behavioral interventions like consistent use (reducing transmission by 80-95% in observational data) remain essential, as vaccines in target HIV-specific prevention but not co-occurring sexually transmitted infections. Prospective strategies emphasize adaptive layering, where vaccine rollout would prioritize high-incidence groups alongside scale-up and voluntary medical male circumcision, informed by dynamic models projecting optimal sequencing to minimize new infections amid evolving epidemics. Challenges include potential , though empirical data from implementation show negligible increases in condomless sex (e.g., iPrEx trial substudies reported no significant behavioral disinhibition), suggesting vaccines could similarly integrate without undermining complementary tools. Ongoing trials, such as those evaluating next-generation candidates like mosaic immunogens, incorporate background prevention uptake metrics to assess real-world complementarity, underscoring the need for vaccines conferring partial, broadly neutralizing responses to enhance, rather than supplant, the multifaceted prevention portfolio.

References

  1. [1]
    Progress and Recent Developments in HIV Vaccine Research - NIH
    Jun 26, 2025 · 3.2.​​ The development of an HIV vaccine has faced various challenges, with the most significant obstacles being genetic variability and high ...
  2. [2]
    Safety and immunogenicity of an HIV vaccine trial with DNA prime ...
    Jul 2, 2025 · More than 300 HIV vaccine trials have been conducted, including 11 efficacy trials, with most failing to demonstrate protective immunity.
  3. [3]
    Major Scientific Hurdles in HIV Vaccine Development
    Oct 28, 2020 · In this review, we highlight the history of HIV vaccine development efforts, highlighting major challenges and future directions.
  4. [4]
    Progress and Challenges in HIV-1 Vaccine Research - MDPI
    This review summarizes current approaches utilized to further understand HIV-1 biology and to create a global vaccine.
  5. [5]
    Review Prophylactic HIV-1 vaccine trials: past, present, and future
    This Review discusses lessons learned from clinical trials of HIV-1 vaccines, current technologies that are being explored, and future considerations
  6. [6]
    Biotech's role in advancing HIV vaccine development - PMC - NIH
    Jul 23, 2024 · Yet, the HIV vaccine product development field has not made real progress in 40 years despite advancements in novel vaccine technology and an ...Introduction · Table 1 · Challenges And Opportunities<|control11|><|separator|>
  7. [7]
    News Release: HIV vaccines tested in PrEPVacc fail to reduce ...
    Jul 23, 2024 · MUHAS Newsletter: HIV vaccines in PrEPVacc trial fail to reduce infections: Results announced at AIDS 2024 conference.Missing: successes | Show results with:successes
  8. [8]
    'Devastating': NIH cancels future funding plans for HIV vaccine ...
    May 30, 2025 · Researchers for 40 years have sought to develop an HIV vaccine that could provide years of protection against infection and, before there ...
  9. [9]
    The Question of HIV Vaccine: Why Is a Solution Not Yet Available?
    Apr 8, 2024 · Although many strategies have been designed and tested, an effective anti-HIV-1 vaccine is still not available.
  10. [10]
    History of HIV Vaccine Research | NIAID
    Oct 22, 2018 · 1987. The first HIV vaccine clinical trial opened at the National Institutes of Health (NIH) Clinical Center in Bethesda, Maryland. This Phase 1 ...
  11. [11]
    A brief history of the global effort to develop a preventive HIV vaccine
    Aug 2, 2013 · Perhaps the first attempt to organize a systematic search for an HIV vaccine occurred as early as 1984 when Robert Gallo established an informal ...
  12. [12]
    HIV vaccines 1983–2003 | Nature Medicine
    Jul 1, 2003 · Early trials of T-cell vaccines in humans. Several HIV vaccines have entered phase 1 and 2 clinical trials in uninfected volunteers in the ...
  13. [13]
    HIV Vaccine Development: 35 Years of Experimenting in the ...
    Dec 19, 2020 · In the beginning of HIV vaccine development, the effort was left almost entirely to the private sector, because it was perceived to be ...
  14. [14]
  15. [15]
    Exploring HIV Vaccine Progress in the Pre-Clinical and Clinical Setting
    This review delves into the history of HIV vaccines and the current efforts in HIV prevention, emphasizing pre-clinical vaccine development.
  16. [16]
    Extremely High Mutation Rate of HIV-1 In Vivo - PMC
    Sep 16, 2015 · The high levels of genetic diversity of the HIV-1 virus grant it the ability to escape the immune system, to rapidly evolve drug resistance, and ...Missing: variability | Show results with:variability
  17. [17]
    Innate Immune Evasion Strategies by Human Immunodeficiency ...
    The ability of genetic variability of HIV-1 is one of the major immune evasion strategies for the virus. The HIV-1 RNA genome can be mutated randomly which ...
  18. [18]
    Stochastic Simulations Suggest that HIV-1 Survives Close to Its Error ...
    We estimated the error threshold of HIV-1 to be ∼2–6-fold higher than its natural mutation rate, suggesting that HIV-1 survives close to its error threshold.
  19. [19]
    The external domains of the HIV-1 envelope are a mutational cold spot
    Oct 9, 2015 · We find that the viral mutation rate experiences a threefold reduction in the region encoding the most external domains of the viral envelope.
  20. [20]
    Viral mechanisms of immune evasion - PMC - PubMed Central
    Genetic variability can also generate variant peptide sequences that are either new antigens or that do not bind to major histocompatibility complex (MHC) ...
  21. [21]
    The Route of HIV Escape from Immune Response Targeting Multiple ...
    Antigenic escape is one of the major mechanisms of HIV resilience in face of an active immune response, impeding effective vaccine design [3] and implicated in ...Missing: variability | Show results with:variability
  22. [22]
    HIV Restriction Factors and Mechanisms of Evasion - PubMed Central
    The mechanisms for evasion from restriction factors are virus encoded and frequently involve HIV-1's regulatory/accessory proteins, namely, Vif, Nef, Vpu, and ...Missing: variability | Show results with:variability
  23. [23]
    Major Scientific Hurdles in HIV Vaccine Development - Frontiers
    In this review, we highlight the history of HIV vaccine development efforts, highlighting major challenges and future directions.<|separator|>
  24. [24]
    Animal studies are inaccurate for HIV research - PMC
    Animal models of HIV have been notoriously inaccurate for two reasons. Firstly, the immune response is intensely complicated and there are many disparities.
  25. [25]
    Animal models for HIV/AIDS research | Nature Reviews Microbiology
    Nov 16, 2012 · Animal models offer obvious advantages in the study of HIV/AIDS, allowing for a more invasive investigation of the disease and for preclinical testing of drugs ...
  26. [26]
    Nonhuman primate models and the failure of the Merck HIV-1 ...
    A replication defective adenovirus-type 5 Gag-Pol-Nef HIV-1 vaccine developed by Merck failed in the STEP human Phase IIb efficacy trial.
  27. [27]
    Use of Humanized Mouse Models for Studying HIV-1 Infection ...
    The main obstacle with the BLT mouse model is the laborious surgical manipulation and time–consuming human cell reconstitution required, which leads to higher ...<|control11|><|separator|>
  28. [28]
    The Flaws and Human Harms of Animal Experimentation - PMC - NIH
    HIV/AIDS vaccine research using NHPs represents one of the most notable failures in animal experimentation translation. Immense resources and decades of time ...
  29. [29]
    The quest for a T cell-based immune correlate of protection against ...
    Dec 17, 2010 · The lack of natural immunity against HIV has been a major hindrance to the search for immune correlates of protection.
  30. [30]
    Complex Immune Correlates of Protection in HIV-1 Vaccine Efficacy ...
    This review will discuss recent advances in the identification of immune CoP for HIV-1, with a focus on antibody correlates.
  31. [31]
    Reappraising the Value of HIV-1 Vaccine Correlates of Protection ...
    Apr 6, 2022 · We analyze why so many HIV-1 vaccines have failed, summarize the outcomes of vaccination in NHPs and humans, and discuss the value and pitfalls of hunting for ...
  32. [32]
    Progress and Challenges in HIV-1 Vaccine Research - NIH
    In 1998, the first large-scale, randomized, double-blind, placebo-controlled HIV-1 vaccine trial (VAX004) was launched in the US and Europe. This Phase 3 trial ...
  33. [33]
    Lessons Drawn from Recent HIV Vaccine Efficacy Trials - PMC
    As a welcome surprise to many, the trial demonstrated a 31% reduction in HIV incidence, a marginal but statistically significant result. More than 30 ...
  34. [34]
    First Phase I human clinical trial of a killed whole-HIV-1 vaccine
    Nov 28, 2016 · The complete inactivation of HIV-1 to generate a killed whole-HIV-1 vaccine is the most critical step to develop a killed whole-virus vaccine.
  35. [35]
    Challenges in the development of an HIV-1 vaccine - Nature
    Oct 2, 2008 · To a great extent, HIV-1 vaccine science is still in its infancy. Major unsolved problems remain, and a renewed commitment to basic discovery ...Virologic And Immunologic... · Hiv-1-Specific Cellular... · The Step Study
  36. [36]
    Killed whole-HIV vaccine; employing a well established strategy for ...
    Sep 12, 2017 · However, vaccine development against HIV continues to struggle due to unresolved questions on the exact components of the human immune response ...<|control11|><|separator|>
  37. [37]
    Broadly neutralizing antibodies targeting HIV
    This review discusses recent advancements and obstacles in applying bNAbs and proposes strategies to enhance their therapeutic potential.
  38. [38]
    Probabilities of developing HIV-1 bNAb sequence features ... - Nature
    Nov 6, 2023 · We formally show that lower probabilities for bNAbs are predictive of higher HIV-1 neutralization activity.
  39. [39]
    HIV Vaccine Development at a Crossroads: New B and T Cell ...
    The field has now pivoted to bold novel approaches, including sequential immunization strategies to drive the generation of broadly neutralizing antibodies.
  40. [40]
    The Germline Targeting Vaccine Concept: Overview and Updates ...
    Feb 21, 2025 · Here, we discuss the development of germline-targeting Env immunogens, their in vivo evaluation, and the strategies currently under evaluation.
  41. [41]
    Precise targeting of HIV broadly neutralizing antibody precursors in ...
    May 15, 2025 · A vaccine to prevent infection with HIV should induce broadly neutralizing antibodies (bnAbs) against antigenically diverse virus strains.Missing: novel | Show results with:novel
  42. [42]
    Germline-targeting HIV vaccination induces neutralizing antibodies ...
    Eliciting potent and broad neutralizing antibodies (bnAbs) is a major goal in HIV-1 vaccine development. Here, we describe how germline-targeting immunogen ...
  43. [43]
    A vaccine strategy for inducing broadly neutralizing antibodies ...
    Jun 17, 2024 · Germline-targeting vaccine design holds promise by using immunogens ... targeting HIV vaccines, and a phase I clinical trial has been initiated.
  44. [44]
    Research from the IAVI Neutralizing Antibody Center and partners ...
    May 30, 2024 · IAVI's research and development program designed to elicit broadly neutralizing antibodies to HIV is based on a strategy called germline targeting.
  45. [45]
    Strategies For Eliciting Multiple Lineages of Broadly Neutralizing ...
    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 ...
  46. [46]
    Vaccination induces HIV broadly neutralizing antibody precursors in ...
    Dec 2, 2022 · Vaccine strategies that induce broadly neutralizing antibodies (bnAbs) have promise to combat HIV and other pathogens but have not yet been tested in humans.
  47. [47]
    Vaccine induction of heterologous HIV-1-neutralizing antibody B cell ...
    May 17, 2024 · This study demonstrates proof of concept for rapid vaccine induction of human B cell lineages with heterologous neutralizing activity and ...
  48. [48]
    Structure‐guided envelope trimer design in HIV‐1 vaccine ... - NIH
    Nov 21, 2021 · This review summarizes the relevant literature focusing on studies using structural biology to reveal and define HIV‐1 Env sites of vulnerability.
  49. [49]
    Design and crystal structure of a native-like HIV-1 envelope trimer ...
    Aug 28, 2017 · Structure-guided design has successfully produced Env-based proteins with increased affinity for gl-bNAbs of the VRC01 class, designated eOD-GT6 ...Design Of The Bg505 Sosip... · Bg505 Sosip. V4. 1-Gt1... · Models Of The Bg505 Sosip...
  50. [50]
    Structure-based Design of Cyclically Permuted HIV-1 gp120 Trimers ...
    The trimeric envelope glycoprotein (Env) spikes on HIV-1 are known to contain multiple epitopes that are susceptible to bNAbs isolated from infected individuals ...
  51. [51]
    An engineered immunogen activates diverse HIV broadly ... - Science
    Jan 8, 2025 · Nevertheless, our work illustrates how controlling the molecular details of antibody-antigen interactions using structure-based design and ...
  52. [52]
    Clinical trials of broadly neutralizing monoclonal antibodies in ...
    Apr 6, 2025 · This review evaluates the clinical outcomes and potential of bNAbs in people living with HIV, summarizing findings from a review of 154 registered trials.Missing: novel | Show results with:novel
  53. [53]
    Two HIV vaccine trials show proof of concept for pathway to broadly ...
    May 15, 2025 · Now, a new study combining data from two separate phase 1 clinical trials shows that a targeted vaccine strategy can successfully activate early ...
  54. [54]
    Two HIV vaccine trials show proof of concept for pathway to broadly ...
    May 15, 2025 · Two phase 1 clinical trials tested the first steps of a germline-targeting HIV vaccine strategy to induce broadly neutralizing antibodies (bnAbs).
  55. [55]
    mRNA vaccines for HIV trigger strong immune response in people
    Aug 1, 2025 · Around 41 million people globally live with HIV, for which there is currently no vaccine. To design vaccines against a virus, researchers often ...
  56. [56]
    Vaccination with mRNA-encoded membrane-anchored HIV ...
    Jul 30, 2025 · mRNA technology might accelerate development of an urgently needed preventive human immunodeficiency virus (HIV) vaccine.
  57. [57]
    Phase 1 clinical trial of HIV vaccine starts in Africa to evaluate ... - IAVI
    Aug 4, 2025 · This Phase 1, first-in-human clinical trial will enroll approximately 120 healthy adults aged 18–50 years, including 48 people living with HIV ...
  58. [58]
    Simultaneous priming of HIV broadly neutralizing antibody ... - Science
    Oct 17, 2025 · The germline-targeting approach for HIV vaccines uses priming envelope (Env) immunogens that activate naïve B cells expressing potentially ...
  59. [59]
    Past, Present, and Future of Viral Vector Vaccine Platforms
    May 15, 2025 · Over the past several decades, viral vector-based vaccines have emerged as some of the most versatile and potent platforms in modern ...
  60. [60]
    Developing the next-generation of adenoviral vector vaccines - PMC
    Jul 1, 2025 · More concerningly, an increased rate of HIV acquisition in men (but not women) was seen after vaccination with HAd5 vectors encoding HIV ...
  61. [61]
    ReiThera Srl, the Ragon Institute and IAVI announce collaboration to ...
    Jan 22, 2024 · A novel HIV vaccine candidate that will be composed of ReiThera's GRAd vector and HIV T-cell epitopes identified by the Ragon Institute.<|separator|>
  62. [62]
    HIV Vaccines - IAVI
    Researchers at the IAVI Vaccine Design and Development Lab are exploring a T-cell vaccine candidate using a recombinant vesicular stomatitis virus (VSV) vector ...
  63. [63]
    Accelerating vaccine development: Plug-and-play platforms for ...
    This study provides insights of “plug-and-play” vaccine platforms, including mRNA, viral-vector, and recombinant protein/nanoparticle chassis, which shift the ...
  64. [64]
    New vector and vaccine platforms: mRNA, DNA, viral vectors - LWW
    In this article, I provide a brief overview of the challenges and opportunities provided by the RNA, DNA and viral vector platforms delivering HIV-1 immunogens.
  65. [65]
    Phase I/II study of a candidate vaccine designed against the B and E ...
    The results suggest that AIDSVAX B/E is safe and immunogenic in humans. The optimal dose for humans in developing countries was 300 microg of each antigen (B ...
  66. [66]
    A Phase I Evaluation of the Safety and Immunogenicity of ...
    Jun 13, 1991 · This gp160 vaccine is safe and immunogenic in volunteer patients with early HIV infection. Although it is too early to know whether this ...
  67. [67]
    A Study of AIDSVAX B/B and AIDSVAX B/E, Two Possible Vaccines ...
    A phase I/II trial to evaluate the safety and immunogenicity of the AIDSVAX B/B vaccine in the United States (final report). 8th Conf Retro and Opportun ...
  68. [68]
    Safety and Reactogenicity of Canarypox ALVAC-HIV (vCP1521) and ...
    In Thailand, phase I/II trials of ALVAC-HIV (vCP1521) and several recombinant gp120 or gp160 boosts showed that the vaccines were well tolerated and immunogenic ...
  69. [69]
    Safety and Immunogenicity of Ad26-Vectored HIV Vaccine With ...
    These findings highlight the potential of Ad26-delivered HIV-1 mosaic antigens combined with recombinant gp140 as vaccine immunogens.
  70. [70]
    Safety and Immunogenicity of a Randomized Phase 1 Prime-Boost ...
    Methods. A phase 1 trial was conducted. In part A (open label), 19 volunteers received oligomeric glycoprotein 160 from HIV strains MN and LAI-2 ( ...
  71. [71]
    Current approaches to HIV vaccine development: a narrative review
    Nov 21, 2021 · The HVTN 137 Phase 1 trial is currently underway evaluating the safety and immunogenicity of BG505 SOSIP.664 with several adjuvants and the ...<|separator|>
  72. [72]
    Safety and immunogenicity of an HIV-1 prefusion-stabilized ...
    Trimer 4571 was the first prefusion-closed stabilized HIV-1 Env trimer to be evaluated in humans and was safe and well tolerated when adjuvanted with alum.
  73. [73]
    Randomized, Double‐Blind, Placebo‐Controlled Efficacy Trial of a ...
    The vaccines in both trials appeared to be safe, and earlier concerns about possible enhancement of HIV‐1 disease progression [31–33 ] could not be confirmed.
  74. [74]
    HIV vaccine immunogenicity and efficacy in clinical trials
    The phase 2b/3 HVTN 702 clinical trial1 constitutes the sixth completed human active HIV immunisation vaccine trial that did not show efficacy. Of these six ...Introduction · Phase 2b/3 Trial Rv144 And... · Phase 1/2 Trial Hvtn 100 And...
  75. [75]
    An effective HIV vaccine: A combination of humoral and ... - PubMed
    RV144, the largest ever HIV vaccine trial, used a prime-boost combination vaccine, which was shown to be safe and modestly effective. The ongoing RV152 study ...
  76. [76]
    RV 144 - an overview | ScienceDirect Topics
    Efficacy trials of six candidate vaccines VAX004, VAX003, Step, Phambili, RV144, and HVTN505 proved RV144 as the only HIV-1 vaccine trial that is effective ...Review Of Efficacy Trials Of... · 4.3. 5 Rv144 · 4.3. 5.3 Efficacy
  77. [77]
    Why did an HIV vaccine fail in the Imbokodo trial? - Aidsmap
    Feb 23, 2022 · Why did an HIV vaccine fail in the Imbokodo trial? Primary tabs. View ... That left just two large HIV vaccine trials underway: Imbokodo and ...Missing: post- PrEPVacc
  78. [78]
    Another HIV vaccine fails to provide protection in discontinued trial
    Jan 18, 2023 · Another HIV vaccine fails to provide protection in discontinued trial ... Both the Mosaico and Imbokodo studies are in follow-up, Wallace said.Missing: PrEPVacc | Show results with:PrEPVacc
  79. [79]
    HIV vaccine research 'at a crossroads' after recent failures - Healio
    Apr 18, 2023 · There are no HIV vaccines currently available, and after the recent failures of three experimental vaccines in development, there are now zero candidates in ...Missing: PrEPVacc | Show results with:PrEPVacc
  80. [80]
    History of CAVD - Collaboration for AIDS Vaccine Discovery
    In the early 2000's the HIV vaccine research community looked inward to find ways to improve the process of vaccine discovery.
  81. [81]
    BioNTech Announces New Collaboration to Develop HIV and ...
    Bill & Melinda Gates Foundation invests $55 million in an infectious disease collaboration that could reach up to $100 million in total funding.
  82. [82]
    Funders - IAVI
    IAVI has been fortunate to receive funding from a diverse array of supporters, including government agencies, philanthropic organizations, and academic ...
  83. [83]
    Global HIV Vaccine Enterprise | International AIDS Society (IAS)
    The Global HIV Vaccine Enterprise promotes HIV vaccine development by sharing knowledge, fostering collaboration, prioritizing HIV vaccine R&D on the global ...
  84. [84]
    Investment by Technology - HIV Resource Tracking Working Group
    Overall European investment in preventive HIV vaccine R&D increased by 47 percent and amounted to US$27.5 million. Philanthropic contributions decreased from US ...Missing: 2020-2025 | Show results with:2020-2025
  85. [85]
    Funding Trends - HIV Resource Tracking Working Group
    In 2021, total funding for HIV prevention R&D increased 12.5 percent (US$140 million) from the previous year, rising to US$1.25 billion.Missing: 2020-2025 | Show results with:2020-2025
  86. [86]
    A promising new HIV vaccine was set to start trials. Then ... - NPR
    Jun 10, 2025 · That work was funded by the largest National Institutes of Health-funded program devoted to such research, with a seven-year grant totaling $258 ...Missing: sources | Show results with:sources<|separator|>
  87. [87]
    HIV vaccine research funding faces an uncertain future - IAVI
    Jun 5, 2025 · The U.S. National Institutes of Health (NIH), which oversees NIAID, is, or at least was, one of the world's largest funders of HIV vaccine ...Missing: sources | Show results with:sources
  88. [88]
    Impact of Cuts to the Consortia for HIV/AIDS Vaccine Development ...
    Jul 1, 2025 · The annual funding for the consortia—approximately $67M—represents a significant chunk of the NIH's funding for HIV vaccine development ...
  89. [89]
    After NIH ends landmark HIV research program, Duke has 9 months ...
    Sep 24, 2025 · The NIH notification and final funding payment started a one-year countdown until CHAVD will no longer receive federal support for its work.
  90. [90]
    NIH ponders overhauling HIV budget to capitalize on prevention ...
    Aug 11, 2025 · Scientists warn $1 billion push for “implementation science” spurred by promise of lenacapavir will hurt research on new treatments and vaccines.
  91. [91]
    HIV Resource Tracking Working Group
    The Working Group's 2021 analysis builds on the US$20 billion in funding tracked between 2000 and 2020 and underscores the importance of continued innovation in ...Missing: total | Show results with:total<|separator|>
  92. [92]
    Trump administration's 'catastrophic' cuts to HIV/AIDS funding ...
    Sep 15, 2025 · In May, the NIH announced it would eliminate the annual $67 million budget for the Consortia for HIV/AIDS Vaccine Development, which has worked ...
  93. [93]
    potential cost‐effectiveness of HIV vaccines and PrEP in Seattle, WA
    Aug 11, 2019 · HIV vaccines are estimated to have an ICER of $42,473/QALY, considered cost‐effective using a threshold of $150,000/QALY. Results were most ...
  94. [94]
    Projected effectiveness and added value of HIV vaccination ... - Nature
    Apr 17, 2018 · Assuming a launch price of $15 per dose, vaccination was found to be cost-effective, with an incremental cost-effectiveness ratio of $13,746 per ...
  95. [95]
    Modeling the economic benefits of an AIDS vaccine - ScienceDirect
    The expected medical savings from a 75% effective vaccine would exceed US$ 25.00 per person for over 700 million people. Although an AIDS vaccine would save ...
  96. [96]
    Do we still need an HIV vaccine now that we have a twice-a-year ...
    Aug 27, 2025 · Even with a powerful new HIV prevention drug, researchers at Fred Hutch say a vaccine is still essential to ending the epidemic.
  97. [97]
    Cheap supplies of HIV-prevention jab for poorer countries hailed as ...
    Sep 24, 2025 · Lenacapavir described as 'next best thing' to an HIV vaccine will cost $40 a year for each patient in 120 countries from 2027, funders say.Missing: opportunity | Show results with:opportunity
  98. [98]
    Excess HIV Infections and Costs Associated With Reductions in ...
    Sep 11, 2025 · Findings This economic evaluation estimated that if PrEP coverage declined modestly—approximately 3% annually—8618 new infections would fail to ...
  99. [99]
    Funding cuts to US HIV preventive services could lead to ... - CIDRAP
    Sep 12, 2025 · ... PrEP locations could reduce PrEP coverage, resulting in excess HIV infections and costs." 8,600 excess HIV infections, $3.6 billion in costs.
  100. [100]
    Why Don't We Have an HIV Vaccine? | Johns Hopkins
    Nov 29, 2022 · The priority would be to get them vaccinated before becoming sexually active to maximize the benefit. I think young adults would be willing to ...
  101. [101]
    [PDF] The world needs an HIV vaccine - IAVI
    And HIV vaccine research has contributed to medical advances for other diseases, leading to longer, healthier lives for people all over the world. No human ...
  102. [102]
    [PDF] Ethical considerations in HIV prevention trials | UNAIDS
    In order to test if an HIV prevention intervention works, large numbers of individuals at high risk of acquiring HIV must be recruited for clinical trials.
  103. [103]
    [PDF] Ethical considerations - HVTN
    Generally, early clinical phases of biomedical HIV prevention research should be conducted in communities that are less vulnerable to harm or. Page 9. Ethical ...
  104. [104]
    Comprehension of informed consent and voluntary participation in ...
    Mar 13, 2024 · This study aimed to find out volunteers' comprehension of informed consent and voluntary participation in Human Immunodeficiency Virus (HIV) clinical trials
  105. [105]
    Placebo use in vaccine trials: Recommendations of a WHO expert ...
    Placebo use in vaccine trials is clearly unacceptable when (a) a highly efficacious and safe vaccine exists and is currently accessible in the public health ...
  106. [106]
    Revised UNAIDS/WHO Ethical Guidance for HIV Prevention Trials
    Jan 27, 2021 · The resulting revised guidance addresses from the perspective of UNAIDS/WHO essential ethical elements for the development of safe and effective HIV prevention ...
  107. [107]
    ethical considerations for including PrEP in a phase IIb HIV vaccine ...
    In fact, UNAIDS' Ethical Considerations in Biomedical HIV Prevention Trials states that investigators should provide all “state of the art” risk reduction ...
  108. [108]
    Descriptions of risks and benefits of HIV vaccine trials in low-and ...
    Jul 3, 2025 · This integrative review aimed to determine the risks and benefits of HIV vaccine trial participation reported in the LMICs from 1995 to 2024.
  109. [109]
    [PDF] Ethics Guidance for Research - HPTN
    HIV prevention researchers should assess, monitor and respond to the social, cultural, and other factors that may place research participants at heightened risk ...
  110. [110]
    [PDF] Ethical Issues in the Design and Conduct of HIV Vaccine Trials
    Others may assume that trial participants are members of groups at increased risk for. HIV infection and social stigmatization could result. Some have suggested ...
  111. [111]
    HIV vaccine raised infection risk - Nature
    Oct 17, 2013 · In the larger of the two trials, called STEP, the vaccine seemed to increase the risk of infection, especially in men. But the risk declined ...
  112. [112]
    Extended Follow-up Confirms Early Vaccine-Enhanced Risk of HIV ...
    The Step Study tested whether an adenovirus serotype 5 (Ad5)–vectored human immunodeficiency virus (HIV) vaccine could prevent HIV acquisition and/or reduce ...
  113. [113]
    STEP analysis confirms higher HIV risk in some vaccine participants
    May 14, 2012 · Ann Duerr follow-up study of experimental HIV vaccine verifies certain subgroups had increased risk of becoming infected after vaccination; risk decreased over ...
  114. [114]
    The failed HIV Merck vaccine study: a step back or a launching point ...
    The scientific community has recently learned that the STEP HIV vaccine trial, which used an rAd5-based vaccine developed by Merck, failed to protect Ad5- ...Missing: 2024 | Show results with:2024
  115. [115]
    STEP study: disappointing, but not a failure - The Lancet
    Not only had the vaccine failed to prevent infection or to reduce viral load, but also further analysis suggested that the vaccine might have actually increased ...
  116. [116]
    HIV Vaccines - Lessons learned and the way forward - PMC - NIH
    An effective HIV vaccine is a global health priority. We describe lessons learned from four HIV vaccine trials that failed to demonstrate efficacy and one ...
  117. [117]
    Pre-Exposure Prophylaxis | HIV.gov
    PrEP is highly effective at preventing HIV when taken as indicated. PrEP reduces the risk of getting HIV from sex by about 99% when taken as prescribed. Among ...HIV Treatment as Prevention · Post-Exposure Prophylaxis
  118. [118]
    Twice-Yearly Lenacapavir or Daily F/TAF for HIV Prevention in ...
    Jul 24, 2024 · Coformulated with emtricitabine, F/TAF is administered in a smaller tablet than F/TDF and is similarly effective for PrEP in cisgender men and ...
  119. [119]
    A Promising Injectable for HIV Prevention - Yale School of Medicine
    Oct 3, 2024 · Lenacapavir showed 96% prevention efficacy compared to background incidence rates and was 89% more effective than the oral drugs. 99.9% of ...
  120. [120]
    PrEP for HIV Prevention: Evidence, Global Scale-up, and Emerging ...
    Apr 8, 2020 · Antiretroviral pre-exposure prophylaxis (PrEP) for the prevention of HIV infection was demonstrated to be efficacious and safe earlier this decade from pivotal ...
  121. [121]
    Does the world still need an HIV vaccine? Experts say yes - Healio
    Nov 27, 2024 · We asked experts during HIVR4P if, given the success of long-acting injectables, there is still a need for an HIV vaccine. They all said yes, ...
  122. [122]
    Key HIV Vaccine Topics - HVTN
    Feb 18, 2025 · A preventive vaccine against the human immunodeficiency virus (HIV), the virus that causes AIDS, is a top priority for human health.
  123. [123]
    Germline-targeting Strategies to Induce bNAbs against HIV-1
    Jun 10, 2025 · This review seeks to synthesize key developments in GT strategies for HIV-1 vaccines, highlighting the de-sign and implementation of immunogens that drive bNAb ...
  124. [124]
    Vaccination induces broadly neutralizing antibody precursors to HIV ...
    May 30, 2024 · Germline-targeting vaccine design posits that bnAbs can be elicited by first priming bnAb precursors with the necessary bnAb-associated genetic ...
  125. [125]
    Is germline targeting the future of HIV vaccine development? | aidsmap
    Jun 12, 2024 · One novel strategy, known as germline targeting, uses a series of primer and booster vaccines to train B cells – the immune system's antibody ...
  126. [126]
    Design and characterization of HIV-1 vaccine candidates to elicit ...
    Aug 12, 2025 · Here, we report a newly designed germline-targeting Env SOSIP trimer, named 3nv.2, that presents three bNAb epitopes on Env: the CD4bs, V3, and ...
  127. [127]
  128. [128]
    Status of HIV vaccine development: progress and promise - PMC - NIH
    May 15, 2025 · ... . FUNDING. All authors were supported by HHS, NIH and NIAID UM1 grant AI144371 for the Consortia for HIV/AIDS Vaccine Development.
  129. [129]
    Combination HIV Prevention Interventions: The Potential of ...
    Combination HIV prevention interventions that integrate efficacious behavioral and biomedical strategies offer the potential to reduce new HIV infections.Missing: tools | Show results with:tools
  130. [130]
    Effectiveness of UNAIDS targets and HIV vaccination across ... - PNAS
    We developed a mathematical model to evaluate the added benefit of an HIV vaccine in the context of goals to increase rates of diagnosis, treatment, and viral ...Abstract · Sign Up For Pnas Alerts · Discussion<|separator|>
  131. [131]
    Full article: Combining biomedical preventions for HIV: Vaccines ...
    From the perspective of controlling the HIV-epidemic, combining a partially effective HIV vaccine with partial effective PrEP, microbicides or other non-vaccine ...Hiv Vaccines · Prevention Of Hiv With... · Case Study Of Prep Use...
  132. [132]
    Finding an AIDS Vaccine in the Context of Combination Prevention
    Sep 18, 2012 · The RV144 vaccine trial in Thailand, which tested a combination of two vaccines, reported a modest 31% efficacy in 2009.