Abstract
Purpose of review
There is growing consensus that eliciting CD8+ T cells in addition to antibodies may be required for an effective HIV vaccine for both prevention and cure. Here, we review key qualities of vaccine-elicited CD8+ T cells as well as major CD8+ T cell-based delivery platforms used in recent HIV vaccine clinical trials.
Recent findings
Much progress has been made in improving HIV immunogen design and delivery platforms to optimize CD8+ T cell responses. With regards to viral vectors, recent trials have tested newer chimp and human adenovirus vectors as well as a CMV vector. DNA vaccine immunogenicity has been increased by delivering the vaccines by electroporation and together with adjuvants as well as administering them as part of a heterologous regimen. In preclinical models, self-amplifying RNA vaccines can generate durable tissue-based CD8+ T cells. While it may be beneficial for HIV vaccines to recapitulate the functional and phenotypic features of HIV-specific CD8+ T cells isolated from elite controllers, most of these features are not routinely measured in HIV vaccine clinical trials.
Summary
Identifying a vaccine capable of generating durable T cell responses that target mutationally vulnerable epitopes and that can rapidly intercept infecting or rebounding virus remains a challenge for HIV. Comprehensive assessment of HIV vaccine-elicited CD8+ T cells, as well as comparisons between different vaccine platforms, will be critical to advance our understanding of how to design better CD8+ T cell-based vaccines for HIV.
Keywords: CD8+ T cell, HIV vaccine, nucleic acid vaccine platforms, T cell quality, viral vectors
INTRODUCTION
The majority of vaccines being developed for HIV prevention aim to elicit antibody responses against the virus, ideally broadly neutralizing antibodies (bNAbs) that can recognize diverse Env sequences [1]. Although there is strong evidence that bNAbs can protect from neutralization-sensitive viral infection in preclinical and clinical studies [2], and while there has been considerable progress towards this goal in recent years, no HIV vaccine strategy to date has successfully generated high titers of HIV bNAbs. T cells, specifically CD8+ T cell responses, can contribute to control of HIV infection [3–5] and therefore may be useful to target in the context of both preventive and therapeutic HIV vaccines. Unlike neutralizing antibodies, virus-specific CD8+ T cells can directly kill infected cells [6]. Additionally, they may offer an added layer of immunity in cases where antibodies are not fully protective [7▪▪,8], they may provide more robust protection against antigen escape (i.e., broader antigen coverage) [9–11], and they may amplify activation and recruitment of other cell types to sites of infection [12].
In this review, we will describe our understanding of ideal features required for HIV vaccine-elicited CD8+ T cells and what is known about the CD8+ T cell immunogenicity of current vaccine platforms that seek to elicit robust virus-specific CD8+ T cell responses. We will not focus on immunogen design, as that has been covered in depth in recent reviews [13,14▪,15▪]. We will discuss methods to comprehensively measure the quality of vaccine-elicited CD8+ T cell responses and, finally, we will consider lessons from HIV therapeutic vaccine studies that may inform prevention strategies.
Box 1.
no caption available
THE ULTIMATE GOAL: WHAT FEATURES DEFINE AN EFFECTIVE HIV-SPECIFIC CD8+ T CELL RESPONSE?
Although most individuals with HIV generate HIV-specific CD8+ T cell responses early in infection [3,5,6,16,17], the majority of people with HIV cannot control viremia without antiretroviral therapy (ART). Rare individuals known as elite controllers [<1% of people with HIV (PWH)] do control viremia to undetectable levels in the absence of ART, and several lines of evidence suggest a role for CD8+ T cells in establishing and maintaining this control [3,4,18▪▪,19–21]. Direct control of infection by CD8+ T cells has been demonstrated by experiments in simian immunodeficiency virus (SIV)- or simian-human immunodeficiency virus (SHIV)-infected nonhuman primates (NHPs) in which CD8α or CD8β depletion led to an increase in viral load [22–27]. Finally, a rhesus cytomegalovirus (RhCMV)-vectored vaccine that elicits CD8+ T cells but no antibody responses has been shown to prevent the establishment of chronic SIV infection in nearly 60% of vaccinated animals [28,29▪▪,30,31▪]. Therefore, CD8+ T cells can, at least in some settings, contribute to control of retroviral infection.
Based on studies in natural HIV/SIV infection and from preclinical testing of HIV vaccine candidates, we believe that successful control of HIV by vaccine-elicited CD8+ T cells will likely require that the CD8+ T cells have the following features (see Fig. 1):
FIGURE 1.
Ideal qualities of vaccine-elicited CD8+ T cell responses (in blue) and assays to comprehensively measure these qualities (purple box). LN, lymph node; TCM, central memory; TCR, T cell receptor; TEM, effector memory.
-
(1)
target viral epitopes that are less likely/unable to be mutated and likely target a broad range of these epitopes across HLA types [13,14▪,15▪,32,33▪],
-
(2)
express T cell receptors (TCRs) with broad epitope reactivity [34,35] and optimal avidity (in some settings, low avidity may enable cross reactivity [36], while in others high avidity may be important for T cell cytotoxic function [34,37]),
-
(3)
are durably maintained at a high magnitude at relevant sites of infection (e.g., gut, rectal, and vaginal mucosa, as well as lymphoid tissue) [18▪▪,38,39], and
-
(4)
occupy a memory-like differentiation state that allows them to robustly proliferate [40▪] and acquire effector functions (e.g., cytotoxicity, cytokine production) upon encountering antigen [20,21,38,41,42].
While many of these features are well defined in the setting of natural HIV infection or preclinical animal models, less is known about how they actually relate to the protective capacity of HIV-specific CD8+ T cells elicited by HIV vaccines administered in people. One clinical trial, HVTN 505 (DNA/Ad5), did report a correlation between Env-specific CD8+ T cell magnitude and polyfunctionality and decreased infection risk (hazard ratio = 0.51 and 0.47, respectively) [43,44]. With regards to epitope targeting, earlier HIV vaccine inserts typically encoded full-length viral proteins, but it is now clear that more narrowly targeting evolutionarily conserved and/or structurally constrained epitopes/regions more efficiently elicits CD8+ T cell responses that are predicted to be less likely to be evaded by viral mutation [13,14▪,15▪,32,33▪,45,46▪▪,47]. Some specific HLA class I alleles have been associated with elite controller status or altered rates of disease progression [19,48–50]. Mamu type-specific effects on vaccine protection have been observed in NHPs [51,52] and HLA-adaptation of T cell epitopes may impact vaccine-elicited T cell responses in people [53▪▪]. In terms of differentiation state, it is unclear which specific differentiation state(s) will be most beneficial/critical to elicit in the context of a preventive vaccine for HIV. Virus-specific memory CD8+ T cells in elite controllers express high levels of the T cell memory-associated transcription factor, TCF-1, and are highly proliferative upon antigen encounter [40▪,54,55]. On the other hand, SIV-specific MHC-E restricted CD8+ T cells with an effector memory phenotype are the predominant subset elicited by protective RhCMV-vectored vaccines [29▪▪,30,31▪,56▪]. As we discuss in the latter section of this review, comprehensive evaluation of all of the HIV-specific CD8+ T cell properties depicted in Fig. 1 will be required to meaningfully compare how different CD8+ T cell-based HIV vaccine platforms elicit them and how they in turn relate to immune protection.
T CELL-BASED VACCINE DELIVERY PLATFORMS
The choice of vaccine delivery platform (e.g., protein, nucleic acid, viral vector) and route of administration determines how immunogens are presented, in what tissues, and for how long, and thus significantly impacts the immunogenicity and durability of vaccine-elicited immune responses [57,58,59▪]. Vaccine platforms that use protein/subunit (AIDSVAX), viral vector (Ad5, ALVAC-HIV, Ad26, MVA), and plasmid DNA (DNA-HIV-PT123, VRC-HIVDNA009-00-VP) have been used in HIV vaccine efficacy trials. Because viral vector and nucleic acid-based delivery platforms can elicit robust T cell responses (unlike protein-based vaccines) [60], we will review what is known about the antigen-specific CD8+ T cell responses elicited by these different vaccine approaches based on recent human HIV vaccine clinical trials in HIV (see Table 1) and other contexts.
Table 1.
Summary of HIV preventive vaccine strategies aimed at eliciting CD8+ T cells
| Year results published | Trial name | NCT # | Phase | Delievery | Vaccine | Immunogen design | % with CD8 response | Notes | Publications; trials (cure) |
| 2008 | HVTN 502, STEP, Merck 023 | NCT00095576 | 2 | Ad5, IM | MRKAd5 HIV-1 | Full sequence consensus; Gag/Pol/Nef (clade B) | 73% | 4 w post-boost; higher rates if low Ad5 titers | [65,67] |
| 2011 | HVTN 204 | NCT00125970 | 2 | Prime: DNA, IM/EP Boost: Ad5, IM |
VRC DNA/rAd5 | Full sequence consensus; Gag/Pol/Nef (clade B) and Env (clades A, B, C) | 47% | 6 w post-boost | [134] Cure: [135] |
| 2013 | HVTN 505 | NCT00865566 | 2 | Prime: DNA, IM/biojector Boost: Ad5, IM |
VRC DNA/rAd5 | Full sequence consensus; Gag/Pol/Nef (clade B) and Env (clades A, B, C) | 64% | 4w post-boost | [83] Cure: [135] |
| 2013 | HVTN 080, PENNVAX | NCT00991354 | 1 | DNA, IM/EP | PENNVAX®-GP HIV-1 DNA vaccine +/- IL-12 DNA | Full sequence consensus; Gag, Pol, and Env (clade B) | 52% | 2w post-3rd dose (+IL-12+EP group) |
[136] Cure: [133], NCT03606213 |
| 2014 | HIV-CORE-002 | NCT01151319 | 1 | Prime: ChAdV63/Boost: MVA; IM. Prime: DNA/Boost: ChAdV62, MVA; IM. Prime: DNA/Boost: MVA, ChAdV62; IM | ChAdV63.HIVconsv, pSG2.HIVconsv, MVA.HIVconsv | Conserved region/consensus sequence; Gag (clades A, C, D), Pol (clades A, B, C, D), Vif (clade D), Env (clades C, D) | response rates not reported | n/a | [13,47,114] Cure (immunogen): [137–140] |
| 2017 | HVTN 087 | NCT01578889 | 1 | Prime: DNA, IM/EP Boost: VSV, IM |
ProfectusVax: HIV-MAG +/- IL-12 DNA + VSV-Gag | Full sequence consensus; Gag/Pol/Nef/Tat/Vif/Env (clade B) | 49% | 6 m post-boost; all participants +IL-12 | [108,112] Cure (HIV-MAG): [141] |
| 2019 | HVTN 098, PENNVAX | NCT02431767 | 1 | DNA, ID or IM [EP] | PENNVAX®-GP HIV-1 DNA vaccine +/- IL-12 DNA | Full sequence consensus; Gag (clades A, B, C, D), Pol, and Env (clades A, C) | 65% (ID), 54% (IM) | 6 m post-boost; +IL-12 group (lower without) | [109▪▪,142] Cure: [133], NCT03606213 |
| 2020 | HVTN 117, TRAVERSE | NCT02788045 | 1/2 | Prime: Ad26, IM Boost: protein, IM |
Ad26.Mos.HIV+Clade C gp140; Ad26.Mos4.HIV+Clade C gp140 | Mosaic; Gag/Pol (based on group M), and Env (clades B, C, CRF01_AE; for Mos4) |
33% | 6 m post-boost; Gag-specific (tetravalent) | [91] Cure (MVA boost): [143] |
| 2021 | HVTN 106 | NCT02296541 | 1 | Prime: DNA, IM Boost: MVA, IM |
DNA Nat-B env or DNA CON-S env or DNA mosaic env plus MVA-CMDR boost | Natural isolate, consensus or mosaic; All express: gp160 Env Nat-B (Clade B), Con-S and mosaic: Env for group M; MVA-CMDR: Env/Gag/Pol (clades A and E) | 29% (Nat-B), 36% (Con-S), 22% (mosaic) | 6m post-boost | [46▪▪,144] |
| 2023 | HVTN 118, ASCENT | NCT02935686 | 1/2 | Prime: Ad26, IM Boost: protein, IM |
Ad26.Mos4.HIV+Clade C gp140; Ad26.Mos4.HIV+Clade C gp140 +Mosaic gp140 | Mosaic; Gag/Pol (group M), and Env (clades B, C, CRF01_AE); Mosaic gp140 (group M) | 18% | 6m post-boost; Gag-specific (mosaic group) | [145▪] Cure (Ad26.Mos4): NCT04983030 |
| 2023 | HVTN 112 | NCT02654080 | 1 | Prime: DNA, IM/EP Boost: VSV, IM |
HIV-1 nef/tat/vif, env pDNA vaccine + rVSV HIV envC | Natural isolate; Nef/Tat/Vif (clade B) and Env (clades B and C) | 18%, 0% | 2w post-boost (1st, 2nd); Env-specific | [146] |
| 2023 (halted) | HVTN 706, Mosaico | NCT03964415 | 3 | Prime: Ad26, IM Boost: protein, IM |
Ad26.Mos4.HIV + Clade C gp140 + Mosaic gp140 | Mosaic; Gag/Pol (group M), and Env (clades B, C, CRF01_AE); Mosaic gp140 (group M) | Cure (Ad26.Mos4): NCT04983030 | ||
| TBD | HIV-CORE-006 | NCT04553016 | 1 | Prime: ChAdOx1, IM; Boost: MVA, IM | ChAdOx1.tHIVconsv1, MVA.tHIVconsv3, MVA.tHIVconsv4 | Conserved/mosaic; Gag/Pol (group M) | Immunogen: [47,114] Cure (immunogen): [147], NCT03844386 |
||
| TBD | HIV-CORE-0051 | NCT04563377 | 1/2a | Prime: ChAdOx1, IM; Boost: MVA, IM | ChAdOx1.HTI, MVA.HTI | T cell responses associated with viral control in PWH; Gag, Pol, Nef, Vif (clades B and C) | Preclinical: [14▪,32] Cure: [129,148], NCT04364035 |
||
| TBD | VIR-1111 | NCT04725877 | 1 | Human CMV, SC | VIR-1111 | UD | |||
| TBD | HVTN 119 | NCT03181789 | 1 | DNA, IM/EP | p24CE1/2 pDNA + p55^gag +IL-12 DNA | Conserved elements; Gag p24, p55 (group M) | Preclinical: [149] Cure: NCT03560258 |
EP, electroporation; ID, intradermal; IM, intramuscular; SC, subcutaneous; UD, undisclosed.
Viral vector vaccines
Viral vectors have been a consistent part of the HIV vaccine pipeline including in the RV144 trial [61–64], designed to elicit antibody responses, and STEP/Phambili trials, designed to elicit CD8+ T cell responses [65–67]. Viral vectors can generate durable T cell responses without the need for an adjuvant [68,69] and can be administered intranasally and orally to specifically target mucosal responses [68,70–74]. Recent and currently active HIV preventive vaccine trials utilize poxvirus viral vectors [modified vaccinia virus Ankara (MVA)], human (Ad4, Ad26) and chimp (AdC6, AdC7, ChAdOx1) adenoviruses, and human cytomegalovirus. Additional viral vectors have been used in other vaccine settings, with the most detailed description of the magnitude, durability, and memory-like qualities of the response being described for the live-attenuated Yellow Fever Vaccine [75–77].
In general, human adenovirus vectors can elicit robust CD8+ T cell responses [68,69]. The human adenovirus vector, Ad5, was the first viral vector to be tested in efficacy trials for HIV (STEP trial/MRKAd-5 HIV), specifically with the goal of eliciting CD8+ T cell responses that target Gag/Pol/Nef [65]. In this trial, nearly 75% of vaccinated participants tested formed detectable HIV-specific T cell responses in response to vaccination as measured by interferon gamma (IFNγ) ELISpot 4 weeks after the last dose [67]. Although the vaccine did not generally elicit a broad CD8+ T cell response [78] and was not protective (vaccinated men who were Ad5 seropositive and uncircumcised had transient increased rates of infection [65,67]), there was an association between vaccine-generated responses to three or more Gag epitopes and reduced viral loads [43]. Much follow-up work has been done to understand the increased risk and overall outcomes of the STEP trial [53▪▪,79,80]. Ad5 continues to be used in heterologous vaccine approaches [81–85]. Other human adenoviruses, Ad26 and Ad35, have also been used due to lower preexisting immunity [86–88]. Preclinical studies in the context of HIV and other settings demonstrate that, compared with Ad5, these vectors generate CD8+ T cell responses at lower magnitude [68,69,86–89], but they may generate responses with improved T cell memory properties (e.g., long-lived Ad26-elicited CD8+ T cells have a more terminally phenotype compared to Ad5-elicited T cells) [87–90]. Ad26 expressing mosaic Gag/Pol/Env immunogens with bivalent Env (clade C/mosaic gp140) protein boost was recently tested in the Mosaico phase III trial (HVTN 706/NCT03964415). Previous trials that utilized earlier iterations of the vaccines used in Mosaico elicited Gag-specific CD8+ T cell responses in 32% (tetravalent [Gag/Pol/Env1/Env2] Ad26 mosaic design) 6 months after the last dose [91]. Mosaico was stopped in early 2023 due to lack of efficacy at preventing HIV infection.
Chimp adenovirus vectors have also been developed to avoid preexisting vector immunity to human adenovirus vectors [92,93] and two chimp adenovirus vectors, ChAdOx1 and AdC6/AdC7, are currently being utilized in phase I clinical trials for HIV (via intramuscular injection; NCT04553016, NCT05182125). In a side-by-side comparisons of chimp to human adenovirus vectors in mice, human Ad5 and chimp Ad3 showed equivalent Gag-specific CD8+ T cell response magnitude (as measured by MHC class I tetramer staining) and protective capacity upon challenge with Listeria monocytogenes expressing SIV Gag [87]. HIV-CORE-002 examined the use of heterologous combinations of ChAdOx63, DNA, and MVA to deliver the Gag/Pol/Vif/Env-containing HIVconsv immunogen in volunteers without HIV and found that 100% of participants generated HIVconsv-specific T cell responses following boost as detected by IFNγ ELISpot for all heterologous vaccine schedules tested [47]. Although relatively new to the HIV vaccine pipeline (HIV-CORE-006, HIV-CORE-051), the ChAdOx1 vector developed by Oxford University/AstraZeneca has recently been widely tested and deployed for SARS-CoV-2 (AZD1222) [94]. After a single dose of the ChAdOx1 vaccine, SARS-CoV-2-specific CD8+ T cells expressing any combination of the cytokines IFNγ, IL-2, and/or TNFα, as identified by intracellular cytokine staining (ICS), were present at approximately 0.1% of total CD8+ T cells 14 days following the vaccine [95]. Compared with lipid nanoparticle (LNP)-formulated mRNA or heterologous (mRNA+ChAdOx1) vaccine approaches, two doses of the ChAdOx1 vaccine elicited a lower overall magnitude of total T cell responses as measured by IFNγ ELISpot [96▪▪,97,98].
The first phase I trial using a human CMV (hCMV) viral vector was recently completed by Vir Biotechnology (NCT04725877), with initial reports indicating that the vaccine is well tolerated [99]. There are several potential advantages of using a CMV vector-based platform to elicit HIV-specific CD8+ T cell responses [29▪▪]. First, based on extensive work on rhCMV strain RhCMV68-1, vaccines with RhCMV68-1 expressing SIV immunogens elicited high magnitude, broad effector memory (TEM)-skewed CD8+ T cell responses in the absence of an antibody response in 100% of animals, and demonstrated arrest and clearance of SIV in nearly 60% of vaccinated rhesus macaques, with similar efficacy maintained in CMV seropositive animals [28,29▪▪,30,31▪,56▪,100]. Second, the RhCMV68-1 vaccine generates unconventional MHC-E-restricted HIV-specific CD8+ T cells [31▪,56▪,101]. MHC-E is highly conserved and has limited polymorphism compared to classical MHC-I, thus potentially increasing the likelihood that conserved epitopes could be found when adapting the CMV platform for use in humans [29▪▪,102]. One outstanding question is whether a human CMV vector containing HIV immunogens has the same capacity to generate unconventional MHC-E-restricted responses, and, ultimately whether these responses can prevent the establishment of chronic HIV infection in humans. Furthermore, while MHC-E-restricted responses can be primed in vitro[103], it is unknown how they may synergize with conventional MHC class I-restricted CD8+ T cell responses and/or other cell types in mediating protection.
Nucleic acid based vaccines
Nucleic acid-based delivery systems (DNA and RNA) offer distinct advantages over viral vectors: they are less expensive and easier to design/manufacture and they circumvent issues with vector immunity and vector backbone immunogenicity [59▪,60,104]. Whereas hundreds of millions of doses of mRNA vaccines for SARS-CoV-2 have now been administered in humans, DNA vaccines remain in more limited use, despite extensive testing in clinical trials for both cancer and HIV [60,105,106].
Since the time of the first clinical trial to test a DNA vaccine in humans (an HIV therapeutic vaccine) [107], the immunogenicity of DNA-based vaccines has improved with delivery via electroporation and design of regimens that include boosting with a viral vector [104,108,109▪▪,110–113]. Using inserts targeting Gag and Pol consensus sequences, the PENNVAX-GP DNA vaccine (HVTN 098) demonstrated the ability of a DNA vaccine alone [delivered via intramuscular (i.m.) or intradermal injection with plasmid IL-12 adjuvant] to elicit CD4+ (96%) and CD8+ (44% i.m., 64% intradermal) T cell responses as well as antibody responses (14% i.m., 56% intradermal) 2 weeks after the final dose [109▪▪]. When comparing different delivery platforms/vaccination schedules utilizing the HIVconsv vaccine insert, DNA prime plus ChAdV63/MVA boost compared with ChAdV63 prime plus MVA, all vaccinees from both vaccine schedules maintained T cell responses as detected by ELISpot two years postvaccination and the magnitude of these responses was not significantly different between the two vaccine schedules [13,114].
mRNA/LNP-based vaccines saw widespread administration for SARS-CoV-2 and two active phase 1 trials are examining the ability of mRNA vaccines to generate bNAbs to HIV Env (NCT05217641, NCT05001373). In the context of SARS-CoV-2, mRNA/LNP vaccinees elicit memory CD8+ T cell responses in approximately 40–60% of vaccinees 6 months after the second dose [11,115,116], and Spike-specific CD8+ T cells are predominantly TEM phenotype, although a stable pool of polyfunctional stem-like memory cells (CD45RA+ CD27+ CD28+ CCR7+ CD95+) with high proliferative capacity can also be detected at long as 9 months after the second dose [11,117▪,118▪▪,119]. For individuals who were vaccinated with mRNA/LNP or ChAdOx1 and who subsequently experienced breakthrough infection, the frequency of activated SARS-CoV-2 Spike-specific CD8+ T cells at symptom onset inversely correlated with viral clearance [118▪▪]. In addition to SARS-CoV-2 vaccines, cancer therapeutic vaccines have specifically sought to optimize CD8+ T cell responses using mRNA platforms [120,121]). Recent preclinical studies are utilizing mRNA as a heterologous boost with DNA [122], and self-amplifying RNA (saRNA) [123▪▪] and circular RNA [124] also demonstrate the potential of RNA-based platforms in eliciting CD8+ T cell responses. Specifically, saRNA delivery of the tHIVconsvX immunogen generated both effector and central memory phenotype CD8+ T cells responses that maintained polyfunctionality and proliferative capacity for 22 weeks postvaccination in mice [125], suggesting that this platform may be an effective approach to improving the durability of tissue-localized responses.
LABORATORY ASSESSMENT OF VACCINE-ELICITED CD8+ T CELLS
Aside from what we have discussed above, relatively little is known about how different vaccine approaches (for HIV or in other contexts) influence the quality of the vaccine-elicited T cell responses on people. This gap in our knowledge exists for many reasons, including the fact that very few controlled studies have been designed to test different vectors [47,126,127], adjuvants [128], and/or immunogens [46▪▪] side-by-side in well matched populations of study participants, and, in general, T cell-based assays, which require viably cryopreserved peripheral blood cells, are more labor and resource-intensive and can be more complex to interpret due to global HLA diversity. In order to address this gap, HIV vaccine trials would ideally measure and report the key features that define the quality of an HIV-specific T cell response (Fig. 1).
Of all these features, assessing T cell proliferative capacity and the ability to sustain killing of target cells may be the highest yield, as these qualities have been the most reliably associated with control in natural infection [21,41,42,54]. Beyond characterizing proliferation and killing capacity, key features of vaccine-elicited CD8+ T cells can be measured by performing deep phenotyping of vaccine-elicited HIV-specific CD8+ T cells by intracellular cytokine staining (ICS) and/or of MHC class I multimer staining by high-dimensional phenotyping and in-situ characterization of tissue-based vaccine-elicited CD8+ T cell responses. Furthermore, integrated systems immunologic assessments of cellular and plasma-based broad immune responses to different vaccine delivery systems can provide insight into the mechanisms by which each vaccine platform promotes the formation of CD8+ T cell responses. Capturing this comprehensive picture of vaccine-elicited CD8+ T cells would allow for a deeper understanding of what type of T cell response each vaccine approach can elicit, it would enable much-needed cross-platform comparisons, and it would also potentially allow for the discovery of novel correlates of protection.
LESSONS FROM HIV CURE STUDIES
While historically most preventive vaccine approaches for HIV have focused on eliciting antibody responses, CD8+ T cell-based vaccines have been a more central focus of HIV cure efforts due to their potential to elicit an immune response capable of clearing established infection. Most of the qualities desired for a preventive vaccine are similar to those desired in the cure setting (e.g., high magnitude and breadth, robust proliferative and killing capacity). Although mucosal-based immune responses may be more important for prevention and lymphoid tissue-based responses are essential for cure, because HIV disseminates so rapidly across lymphoid tissues in the body after infection, preventive vaccines will also need to elicit immune responses that have the capacity to eliminate infected cells in these tissues. Similarly, therapeutic vaccines would also ideally prevent re-infection, and thus should elicit strong immunity at mucosal barriers.
Recent advances in developing CD8+ T cell-based vaccines for HIV cure have been extensively reviewed recently elsewhere [13,14▪,15▪,33▪], and vaccine designs being tested in both the prevention and cure settings are noted on Table 1. A recent study using a heterologous approach with DNA, MVA, ChAd vaccinations and a conserved mosaic insert given to people living with HIV on suppressive ART (AELIX-002) demonstrated robust T cell immunogenicity and a relationship between T cell responses and lower viral loads after ART was discontinued [129]. Data being generated from ongoing therapeutic vaccine studies with vaccines given alone or in combination with other immunotherapeutic modalities, and often with the inclusion of an ART treatment interruption, will therefore directly inform the design of studies for prevention.
COMBINING B AND T CELL RESPONSES
As discussed at a recent NIH-sponsored meeting on ‘T and B cell synergy for HIV vaccines’, an effective vaccine strategy to prevent and/or cure HIV infection will likely require induction of both an effective antibody response (i.e., bNAbs elicited and maintained at a high titer) as well as a potent CD8+ T cell response. To achieve optimal B cell and CD8+ T cell responses, a heterologous approach may be required [7▪▪,130]. Most HIV vaccine approaches described above and listed in Table 1 do not elicit both antibodies and CD8+ T cell responses at a high magnitude/breadth/durability. This is in part due to the different cytokines likely required for optimal germinal center versus memory CD8+ T cell differentiation (i.e., IL-4 versus IL-12/IFNγ, respectively) [131]. In addition, immunogens designed to elicit Env-specific antibody responses may stimulate less effective T cell responses that target nonconserved T cell epitopes. For example, in both a prevention and therapeutic vaccine setting, inclusion of Env sequences has been shown to impair the generation of T cell responses against more conserved regions in Gag and Pol [132▪▪,133]. Going forward, it will be critical to design carefully controlled studies in humans and animal models to systematically evaluate the additive effects and trade-offs of altering vaccine platform or immunogen on the quality of both the antibody and CD8+ T cell response in order to understand how to elicit optimal responses in both arms.
CONCLUSION
In recent years, newer vaccine platforms aimed at eliciting robust CD8+ T cell responses have been tested in the context of HIV, SARS-CoV-2, and cancer, in both preclinical and clinical settings. Going forward, we believe that addressing the following outstanding questions will be critical to move us closer to finding an optimal CD8+ T cell-based vaccine design for HIV prevention and/or cure:
-
(1)
How does vaccine delivery system influence key qualities of the HIV-specific CD8+ T cell responses, such as magnitude (across diverse HLA types), durability, breadth of overall response and specific TCR epitope recognition, TCR avidity, polyfunctionality, proliferative and killing capacity, and homing potential?
-
(2)
Is there a minimum breadth/number of T cell responses required to provide protection? How does immunogen design (and HLA background) affect this number?
-
(3)
Can a single vaccine elicit mucosal-based T cell immunity and also minimize recruitment of activated CD4+ T cells that may be prime target cells for HIV infection?
-
(4)
Can antibody and T cell responses synergize with one another, and are different vaccine platforms and inserts required to elicit optimal antibody versus T cell responses?
Acknowledgements
Figure 1 was created with BioRender.com. Research reported in this publication was supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under R01AI170239, P01AI78375, and UM1AI164560 (R.L.R.), T32AI060530 (G.M.B.), and from the Bill and Melinda Gates Foundation (INV-046661, R.L.R.).
Financial support and sponsorship
None.
Conflicts of interest
There are 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Walsh SR, Seaman MS. Broadly neutralizing antibodies for HIV-1 prevention. Front Immunol 2021; 12:712122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Collins DR, Gaiha GD, Walker BD. CD8+ T cells in HIV control, cure and prevention. Nat Rev Immunol 2020; 20:471–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Takata H, Buranapraditkun S, Kessing C, et al. Delayed differentiation of potent effector CD8+ T cells reducing viremia and reservoir seeding in acute HIV infection. Sci Transl Med 2017; 9:eaag1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ndhlovu ZM, Kamya P, Mewalal N, et al. Magnitude and kinetics of CD8+ T cell activation during hyperacute HIV infection impact viral set point. Immunity 2015; 43:591–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Actor JK. T lymphocytes: ringleaders of adaptive immune function. Introductory immunology [Internet]. Elsevier; 2019 [cited 28 June 2023]. 45-62. https://linkinghub.elsevier.com/retrieve/pii/B9780128165720000048. [Google Scholar]
- 7▪▪.Arunachalam PS, Charles TP, Joag V, et al. T cell-inducing vaccine durably prevents mucosal SHIV infection even with lower neutralizing antibody titers. Nat Med 2020; 26:932–940. [DOI] [PMC free article] [PubMed] [Google Scholar]; Using a heterologous viral vector regimen, this study demonstrates that vaccine-elicited CD8+ T cells can lower the threshold of neutralizing antibodies needed for protection, providing proof-of-concept that vaccine-elicited B and T cell responses can work together to provide durable protection from infection.
- 8.Petitdemange C, Kasturi SP, Kozlowski PA, et al. Vaccine induction of antibodies and tissue-resident CD8+ T cells enhances protection against mucosal SHIV-infection in young macaques. JCI Insight 2019; 4:e126047.126047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gao Y, Cai C, Grifoni A, et al. Ancestral SARS-CoV-2-specific T cells cross-recognize the Omicron variant. Nat Med 2022; 28:472–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Geers D, Shamier MC, Bogers S, et al. SARS-CoV-2 variants of concern partially escape humoral but not T-cell responses in COVID-19 convalescent donors and vaccinees. Sci Immunol 2021; 6:eabj1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tarke A, Coelho CH, Zhang Z, et al. SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron. Cell 2022; 185:847–859. e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Rosato PC, Lotfi-Emran S, Joag V, et al. Tissue-resident memory T cells trigger rapid exudation and local antibody accumulation. Mucosal Immunol 2023; 16:17–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hanke T. Aiming for protective T-cell responses: a focus on the first generation conserved-region HIVconsv vaccines in preventive and therapeutic clinical trials. Expert Rev Vaccines 2019; 18:1029–1041. [DOI] [PubMed] [Google Scholar]
- 14▪.Brander C, Hartigan-O’Connor D. HIV T-cell immunogen design and delivery. Curr Opin HIV AIDS 2022; 17:333–337. [DOI] [PMC free article] [PubMed] [Google Scholar]; A review on different approaches to HIV vaccine immunogen design (e.g., based on correlation with functional control in natural infection, conservation, structurally constrained/networked).
- 15▪.Kaseke C, Tano-Menka R, Senjobe F, Gaiha GD. The emerging role for CTL epitope specificity in HIV cure efforts. J Infect Dis 2021; 223: (Suppl 1): S32–S37. [DOI] [PMC free article] [PubMed] [Google Scholar]; A review on the role of CD8+ T cell epitope specificity and elite control and how the concept of networked epitopes can be applied to HIV vaccine design.
- 16.Demers KR, Makedonas G, Buggert M, et al. Temporal dynamics of CD8+ T cell effector responses during primary HIV infection. PLoS Pathog 2016; 12:e1005805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Borrow P, Lewicki H, Wei X, et al. Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid selection of CTL escape virus. Nat Med 1997; 3:205–211. [DOI] [PubMed] [Google Scholar]
- 18▪▪.Collins DR, Hitschfel J, Urbach JM, et al. Cytolytic CD8+ T cells infiltrate germinal centers to limit ongoing HIV replication in spontaneous controller lymph nodes. Sci Immunol 2023; 8:eade5872. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study characterizes CD8+ T cell responses in lymph nodes, an important site of the HIV reservoir, and finds that HIV-specific CD8+ T cells from HIV controllers are distinguished by their proliferative capacity and ability to differentiate, in the lymph node, into cytotoxic cells in close proximity to cells harboring transcriptionally active HIV.
- 19.Pereyra F, Jia X, McLaren PJ, et al. International HIV Controllers Study. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 2010; 330:1551–1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rutishauser RL, Trautmann L. CD8 + T-cell responses in HIV controllers: potential implications for novel HIV remission strategies. Curr Opin HIV AIDS 2022; 17:315–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ndhlovu ZM, Chibnik LB, Proudfoot J, et al. High-dimensional immunomonitoring models of HIV-1-specific CD8 T-cell responses accurately identify subjects achieving spontaneous viral control. Blood 2013; 121:801–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chowdhury A, Hayes TL, Bosinger SE, et al. Differential impact of in vivo CD8+ T lymphocyte depletion in controller versus progressor simian immunodeficiency virus-infected macaques. J Virol 2015; 89:8677–8686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jin X, Bauer DE, Tuttleton SE, et al. Dramatic rise in plasma viremia after CD8(+) T cell depletion in simian immunodeficiency virus-infected macaques. J Exp Med 1999; 189:991–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cartwright EK, Spicer L, Smith SA, et al. CD8(+) lymphocytes are required for maintaining viral suppression in SIV-infected macaques treated with short-term antiretroviral therapy. Immunity 2016; 45:656–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Nishimura Y, Donau OK, Dias J, et al. Immunotherapy during the acute SHIV infection of macaques confers long-term suppression of viremia. J Exp Med 2021; 218:e20201214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nishimura Y, Gautam R, Chun TW, et al. Early antibody therapy can induce long-lasting immunity to SHIV. Nature 2017; 543:559–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Okoye AA, Duell DD, Fukazawa Y, et al. CD8+ T cells fail to limit SIV reactivation following ART withdrawal until after viral amplification. J Clin Invest 2021; 131:e141677.141677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hansen SG, Vieville C, Whizin N, et al. Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nat Med 2009; 15:293–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29▪▪.Picker LJ, Lifson JD, Gale M, et al. Programming cytomegalovirus as an HIV vaccine. Trends Immunol 2023; 44:287–304. [DOI] [PMC free article] [PubMed] [Google Scholar]; A comprehensive review of the preclinical testing, mechanism(s) of protection, and biology of the rhCMV vaccine vector. It covers open questions related to adapting this platform for a human CMV vectored HIV vaccine.
- 30.Hansen SG, Ford JC, Lewis MS, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 2011; 473:523–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31▪.Hansen SG, Hancock MH, Malouli D, et al. Myeloid cell tropism enables MHC-E-restricted CD8+ T cell priming and vaccine efficacy by the RhCMV/SIV vaccine. Sci Immunol 2022; 7:eabn9301. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that the RhCMV vector tropism determines whether vaccine-elicited CD8+ T cell responses are MHC-Ia, MHC-II, or MHC-E restricted. These results suggest that the CMV vector could potentially be a platform that allows for straightforward modulation of epitope presentation.
- 32.Mothe B, Hu X, Llano A, et al. A human immune data-informed vaccine concept elicits strong and broad T-cell specificities associated with HIV-1 control in mice and macaques. J Transl Med 2015; 13:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33▪.Korber B, Fischer W. T cell-based strategies for HIV-1 vaccines. Hum Vaccines Immunother 2020; 16:713–722. [DOI] [PMC free article] [PubMed] [Google Scholar]; An overview of T cell vaccine design in clinical trials for HIV, with a focus on how different immunogen designs impact breadth and potency of CD8+ T cell responses.
- 34.Chen H, Ndhlovu ZM, Liu D, et al. TCR clonotypes modulate the protective effect of HLA class I molecules in HIV-1 infection. Nat Immunol 2012; 13:691–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ladell K, Hashimoto M, Iglesias MC, et al. A molecular basis for the control of preimmune escape variants by HIV-specific CD8+ T cells. Immunity 2013; 38:425–436. [DOI] [PubMed] [Google Scholar]
- 36.Straub A, Grassmann S, Jarosch S, et al. Recruitment of epitope-specific T cell clones with a low-avidity threshold supports efficacy against mutational escape upon re-infection. Immunity 2023; 56:1269–1284. e6. [DOI] [PubMed] [Google Scholar]
- 37.Almeida JR, Price DA, Papagno L, et al. Superior control of HIV-1 replication by CD8+ T cells is reflected by their avidity, polyfunctionality, and clonal turnover. J Exp Med 2007; 204:2473–2485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Nguyen S, Deleage C, Darko S, et al. Elite control of HIV is associated with distinct functional and transcriptional signatures in lymphoid tissue CD8+ T cells. Sci Transl Med 2019; 11:eaax4077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Buggert M, Nguyen S, Salgado-Montes de Oca G, et al. Identification and characterization of HIV-specific resident memory CD8+ T cells in human lymphoid tissue. Sci Immunol 2018; 3:eaar4526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40▪.Rutishauser RL, Deguit CDT, Hiatt J, et al. TCF-1 regulates HIV-specific CD8+ T cell expansion capacity. JCI Insight 2021; 6:e136648.136648. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates the importance of the transcription factor, TCF-1, and its promotion of stem-like memory qualities, to the superior proliferation of HIV-specific CD8+ T cells from elite controllers.
- 41.Migueles SA, Osborne CM, Royce C, et al. Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity 2008; 29:1009–1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Migueles SA, Rogan DC, Gavil NV, et al. Antigenic restimulation of virus-specific memory CD8+ T cells requires days of lytic protein accumulation for maximal cytotoxic capacity. J Virol 2020; 94:e01595–e1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Janes H, Friedrich DP, Krambrink A, et al. Vaccine-induced gag-specific T cells are associated with reduced viremia after HIV-1 infection. J Infect Dis 2013; 208:1231–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fong Y, Shen X, Ashley VC, et al. Modification of the association between T-cell immune responses and human immunodeficiency virus type 1 infection risk by vaccine-induced antibody responses in the HVTN 505 Trial. J Infect Dis 2018; 217:1280–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Santra S, Liao HX, Zhang R, et al. Mosaic vaccines elicit CD8+ T lymphocyte responses that confer enhanced immune coverage of diverse HIV strains in monkeys. Nat Med 2010; 16:324–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46▪▪.Cohen KW, Fiore-Gartland A, Walsh SR, et al. Trivalent mosaic or consensus HIV immunogens prime humoral and broader cellular immune responses in adults. J Clin Invest 2023; 133:e163338. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study reports results from a phase I trial (HVTN 106) directly comparing antibody and T cell responses from mosaic and consensus immunogen designs delivered with a DNA prime/MVA vector boost and found that mosaic immunogens outperform consensus immunogens in broader epitope recognition that can recognize a limited number of heterologous variants.
- 47.Borthwick N, Ahmed T, Ondondo B, et al. Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther J Am Soc Gene Ther 2014; 22:464–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yu XG, Lichterfeld M, Chetty S, et al. Mutually exclusive T-cell receptor induction and differential susceptibility to human immunodeficiency virus type 1 mutational escape associated with a two-amino-acid difference between HLA class I subtypes. J Virol 2007; 81:1619–1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gaiha GD, Rossin EJ, Urbach J, et al. Structural topology defines protective CD8+ T cell epitopes in the HIV proteome. Science 2019; 364:480–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kløverpris HN, Leslie A, Goulder P. Role of HLA adaptation in HIV evolution. Front Immunol 2015; 6:665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lasaro MO, Haut LH, Zhou X, et al. Vaccine-induced T cells provide partial protection against high-dose rectal SIVmac239 challenge of rhesus macaques. Mol Ther J Am Soc Gene Ther 2011; 19:417–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liang X, Casimiro DR, Schleif WA, et al. Vectored Gag and Env but not Tat show efficacy against simian-human immunodeficiency virus 89.6P challenge in Mamu-A∗01-negative rhesus monkeys. J Virol 2005; 79:12321–12331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53▪▪.Qin K, Boppana S, Carlson JM, et al. Elevated HIV infection of CD4 T cells in MRKAd5 vaccine recipients due to CD8 T cells targeting adapted epitopes. J Virol 2021; 95:e0016021. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study explores the immunological basis of increased viral infection observed in vaccinated participants in the STEP trial. The authors find that HLA-I associated viral adaptation promoted dendritic cell maturation with enhanced ability to facilitate HIV trans-infection.
- 54.Migueles SA, Laborico AC, Shupert WL, et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol 2002; 3:1061–1068. [DOI] [PubMed] [Google Scholar]
- 55.Sekine T, Perez-Potti A, Nguyen S, et al. TOX is expressed by exhausted and polyfunctional human effector memory CD8 + T cells. Sci Immunol 2020; 5:eaba7918. [DOI] [PubMed] [Google Scholar]
- 56▪.Malouli D, Hansen SG, Hancock MH, et al. Cytomegaloviral determinants of CD8+ T cell programming and RhCMV/SIV vaccine efficacy. Sci Immunol 2021; 6:eabg5413. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study reports the genetic determinants of the immunomodulatory components of the RhCMV 68-1 vector and demonstrates that human CMV orthologues are interchangeable with the genes identified in RhCMV 68-1. These findings are directly relevant to the adaptation of RhCMV 68-1 to human CMV for use of the CMV vector in humans.
- 57.Darrah PA, Zeppa JJ, Maiello P, et al. Prevention of tuberculosis in macaques after intravenous BCG immunization. Nature 2020; 577:95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Rosenbaum P, Tchitchek N, Joly C, et al. Vaccine inoculation route modulates early immunity and consequently antigen-specific immune response. Front Immunol 2021; 12:645210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59▪.Ura T, Takeuchi M, Kawagoe T, et al. Current vaccine platforms in enhancing T-cell response. Vaccines 2022; 10:1367. [DOI] [PMC free article] [PubMed] [Google Scholar]; A review covering design aspects of viral vector and mRNA delivery platforms for inducing T cell responses for infectious diseases.
- 60.Hanke T. New vector and vaccine platforms: mRNA, DNA, viral vectors. Curr Opin HIV AIDS 2022; 17:338–344. [DOI] [PubMed] [Google Scholar]
- 61.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. [DOI] [PubMed] [Google Scholar]
- 62.Haynes BF, Gilbert PB, McElrath MJ, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 2012; 366:1275–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kim JH, Excler JL, Michael NL. Lessons from the RV144 Thai phase III HIV-1 vaccine trial and the search for correlates of protection. Annu Rev Med 2015; 66:423–437. [DOI] [PubMed] [Google Scholar]
- 64.Heger E, Schuetz A, Vasan S. HIV Vaccine Efficacy Trials: RV144 and beyond. Adv Exp Med Biol 2018; 1075:3–30. [DOI] [PubMed] [Google Scholar]
- 65.Gray G, Buchbinder S, Duerr A. Overview of STEP and Phambili trial results: two phase IIb test-of-concept studies investigating the efficacy of MRK adenovirus type 5 gag/pol/nef subtype B HIV vaccine. Curr Opin HIV AIDS 2010; 5:357–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Gray GE, Allen M, Moodie Z, et al. Safety and efficacy of the HVTN 503/Phambili study of a clade-B-based HIV-1 vaccine in South Africa: a double-blind, randomised, placebo-controlled test-of-concept phase 2b study. Lancet Infect Dis 2011; 11:507–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Buchbinder SP, Mehrotra DV, Duerr A, et al. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet Lond Engl 2008; 372:1881–1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Travieso T, Li J, Mahesh S, et al. The use of viral vectors in vaccine development. NPJ Vaccines 2022; 7:75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Coughlan L. Factors which contribute to the immunogenicity of nonreplicating adenoviral vectored vaccines. Front Immunol 2020; 11:909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lavelle EC, Ward RW. Mucosal vaccines - fortifying the frontiers. Nat Rev Immunol 2022; 22:236–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Li JX, Hou LH, Gou JB, et al. Safety, immunogenicity and protection of heterologous boost with an aerosolised Ad5-nCoV after two-dose inactivated COVID-19 vaccines in adults: a multicentre, open-label phase 3 trial. Lancet Infect Dis 2023; S1473-3099(23)00350-X. [DOI] [PubMed] [Google Scholar]
- 72.Liebowitz D, Gottlieb K, Kolhatkar NS, et al. Efficacy, immunogenicity, and safety of an oral influenza vaccine: a placebo-controlled and active-controlled phase 2 human challenge study. Lancet Infect Dis 2020; 20:435–444. [DOI] [PubMed] [Google Scholar]
- 73.Liebowitz D, Lindbloom JD, Brandl JR, et al. High titre neutralising antibodies to influenza after oral tablet immunisation: a phase 1, randomised, placebo-controlled trial. Lancet Infect Dis 2015; 15:1041–1048. [DOI] [PubMed] [Google Scholar]
- 74.Jones AT, Shen X, Walter KL, et al. HIV-1 vaccination by needle-free oral injection induces strong mucosal immunity and protects against SHIV challenge. Nat Commun 2019; 10:798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Akondy RS, Fitch M, Edupuganti S, et al. Origin and differentiation of human memory CD8 T cells after vaccination. Nature 2017; 552:362–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Akondy RS, Monson ND, Miller JD, et al. The yellow fever virus vaccine induces a broad and polyfunctional human memory CD8+ T cell response. J Immunol Baltim Md 1950 2009; 183:7919–7930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Fuertes Marraco SA, Soneson C, Cagnon L, et al. Long-lasting stem cell-like memory CD8+ T cells with a naïve-like profile upon yellow fever vaccination. Sci Transl Med 2015; 7:282ra48. [DOI] [PubMed] [Google Scholar]
- 78.Hayes PJ, Cox JH, Coleman AR, et al. Adenovirus-based HIV-1 vaccine candidates tested in efficacy trials elicit CD8+ T cells with limited breadth of HIV-1 inhibition. AIDS Lond Engl 2016; 30:1703–1712. [DOI] [PubMed] [Google Scholar]
- 79.Zak DE, Andersen-Nissen E, Peterson ER, et al. Merck Ad5/HIV induces broad innate immune activation that predicts CD8+ T-cell responses but is attenuated by preexisting Ad5 immunity. Proc Natl Acad Sci U S A 2012; 109:E3503–E3512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Auclair S, Liu F, Niu Q, et al. Distinct susceptibility of HIV vaccine vector-induced CD4 T cells to HIV infection. PLoS Pathog 2018; 14:e1006888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Asmuth DM, Brown EL, DiNubile MJ, et al. Comparative cell-mediated immunogenicity of DNA/DNA, DNA/adenovirus type 5 (Ad5), or Ad5/Ad5 HIV-1 clade B gag vaccine prime-boost regimens. J Infect Dis 2010; 201:132–141. [DOI] [PubMed] [Google Scholar]
- 82.De Rosa SC, Thomas EP, Bui J, et al. HIV-DNA priming alters T cell responses to HIV-adenovirus vaccine even when responses to DNA are undetectable. J Immunol Baltim Md 19502011; 187:3391–3401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hammer SM, Sobieszczyk ME, Janes H, et al. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N Engl J Med 2013; 369:2083–2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Neidich SD, Fong Y, Li SS, et al. Antibody Fc effector functions and IgG3 associate with decreased HIV-1 risk. J Clin Invest 2019; 129:4838–4849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Janes HE, Cohen KW, Frahm N, et al. Higher T-cell responses induced by DNA/rAd5 HIV-1 preventive vaccine are associated with lower HIV-1 infection risk in an efficacy trial. J Infect Dis 2017; 215:1376–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Abbink P, Lemckert AAC, Ewald BA, et al. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. J Virol 2007; 81:4654–4663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Quinn KM, Da Costa A, Yamamoto A, et al. Comparative analysis of the magnitude, quality, phenotype, and protective capacity of simian immunodeficiency virus gag-specific CD8+ T cells following human-, simian-, and chimpanzee-derived recombinant adenoviral vector immunization. J Immunol Baltim Md 1950 2013; 190:2720–2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Tan WG, Jin HT, West EE, et al. Comparative analysis of simian immunodeficiency virus gag-specific effector and memory CD8+ T cells induced by different adenovirus vectors. J Virol 2013; 87:1359–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Penaloza-MacMaster P, Provine NM, Ra J, et al. Alternative serotype adenovirus vaccine vectors elicit memory T cells with enhanced anamnestic capacity compared to Ad5 vectors. J Virol 2013; 87:1373–1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yang TC, Millar J, Groves T, et al. The CD8+ T cell population elicited by recombinant adenovirus displays a novel partially exhausted phenotype associated with prolonged antigen presentation that nonetheless provides long-term immunity. J Immunol Baltim Md 1950 2006; 176:200–210. [DOI] [PubMed] [Google Scholar]
- 91.Baden LR, Stieh DJ, Sarnecki M, et al. Safety and immunogenicity of two heterologous HIV vaccine regimens in healthy, HIV-uninfected adults (TRAVERSE): a randomised, parallel-group, placebo-controlled, double-blind, phase 1/2a study. Lancet HIV 2020; 7:e688–e698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Colloca S, Barnes E, Folgori A, et al. Vaccine vectors derived from a large collection of simian adenoviruses induce potent cellular immunity across multiple species. Sci Transl Med 2012; 4:115ra2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Dicks MDJ, Spencer AJ, Edwards NJ, et al. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity. PLoS One 2012; 7:e40385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sette A, Sidney J, Crotty S. T cell responses to SARS-CoV-2. Annu Rev Immunol 2023; 41:343–373. [DOI] [PubMed] [Google Scholar]
- 95.Ewer KJ, Barrett JR, Belij-Rammerstorfer S, et al. T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nat Med 2021; 27:270–278. [DOI] [PubMed] [Google Scholar]
- 96▪▪.Maringer Y, Nelde A, Schroeder SM, et al. Durable spike-specific T cell responses after different COVID-19 vaccination regimens are not further enhanced by booster vaccination. Sci Immunol 2022; 7:eadd3899. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates the durability and cross-reactivity of T cell responses elicited by a single dose of COVID19 mRNA vaccines, viral vectors, and heterologous vaccination (viral vector/mRNA). T cell responses (both magnitude and overall response rate) and cross recognition of Omicron-variant specific mutations was superior in mRNA and heterologous vaccination compared with viral vector.
- 97.Barros-Martins J, Hammerschmidt SI, Cossmann A, et al. Immune responses against SARS-CoV-2 variants after heterologous and homologous ChAdOx1 nCoV-19/BNT162b2 vaccination. Nat Med 2021; 27:1525–1529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bánki Z, Mateus J, Rössler A, et al. Heterologous ChAdOx1/BNT162b2 vaccination induces stronger immune response than homologous ChAdOx1 vaccination: the pragmatic, multicenter, three-arm, partially randomized HEVACC trial. EBioMedicine 2022; 80:104073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Arvin, A. Vaccine induction of HLA-E mediated protective immunity in humans. NIAID T and B cell synergy for HIV vaccines workshop [virtual]; 2023 Aug 8-9. [Google Scholar]
- 100.Hansen SG, Marshall EE, Malouli D, et al. A live-attenuated RhCMV/SIV vaccine shows long-term efficacy against heterologous SIV challenge. Sci Transl Med 2019; 11:eaaw2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Hansen SG, Womack JL, Perez W, et al. Late gene expression-deficient cytomegalovirus vectors elicit conventional T cells that do not protect against SIV. JCI Insight 2023; 8:e164692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hansen SG, Sacha JB, Hughes CM, et al. Cytomegalovirus vectors violate CD8+ T cell epitope recognition paradigms. Science 2013; 340:1237874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Yang H, Rei M, Brackenridge S, et al. HLA-E-restricted, Gag-specific CD8+ T cells can suppress HIV-1 infection, offering vaccine opportunities. Sci Immunol 2021; 6:eabg1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Xu Z, Patel A, Tursi NJ, et al. Harnessing recent advances in synthetic DNA and electroporation technologies for rapid vaccine development against COVID-19 and other emerging infectious diseases. Front Med Technol 2020; 2:571030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lopes A, Vandermeulen G, Préat V. Cancer DNA vaccines: current preclinical and clinical developments and future perspectives. J Exp Clin Cancer Res CR 2019; 38:146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Suschak JJ, Williams JA, Schmaljohn CS. Advancements in DNA vaccine vectors, nonmechanical delivery methods, and molecular adjuvants to increase immunogenicity. Hum Vaccines Immunother 2017; 13:2837–2848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.MacGregor RR, Boyer JD, Ugen KE, et al. First human trial of a DNA-based vaccine for treatment of human immunodeficiency virus type 1 infection: safety and host response. J Infect Dis 1998; 178:92–100. [DOI] [PubMed] [Google Scholar]
- 108.Elizaga ML, Li SS, Kochar NK, et al. Safety and tolerability of HIV-1 multiantigen pDNA vaccine given with IL-12 plasmid DNA via electroporation, boosted with a recombinant vesicular stomatitis virus HIV Gag vaccine in healthy volunteers in a randomized, controlled clinical trial. PLoS One 2018; 13:e0202753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109▪▪.De Rosa SC, Edupuganti S, Huang Y, et al. Robust antibody and cellular responses induced by DNA-only vaccination for HIV. JCI Insight 2020; 5:e137079.137079. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study reports results from the PENNVAX/HVTN 098 trial investigating the generation of robust T cell responses using DNA. These results demonstrate the cellular immune responses (and antibody responses) can be improved with electroporation and inclusion of plasmid expressing IL-12, with lower doses in the case of intradermal administration.
- 110.Rouphael NG, Morgan C, Li SS, et al. DNA priming and gp120 boosting induces HIV-specific antibodies in a randomized clinical trial. J Clin Invest 2019; 129:4769–4785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Nilsson C, Hejdeman B, Godoy-Ramirez K, et al. HIV-DNA given with or without intradermal electroporation is safe and highly immunogenic in healthy Swedish HIV-1 DNA/MVA vaccinees: a phase I randomized trial. PLoS One 2015; 10:e0131748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Li SS, Kochar NK, Elizaga M, et al. DNA priming increases frequency of T-cell responses to a vesicular stomatitis virus HIV vaccine with specific enhancement of CD8+ T-cell responses by interleukin-12 plasmid DNA. Clin Vaccine Immunol CVI 2017; 24:e00263–e317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Jin X, Morgan C, Yu X, et al. Multiple factors affect immunogenicity of DNA plasmid HIV vaccines in human clinical trials. Vaccine 2015; 33:2347–2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Moyo N, Borthwick NJ, Wee EG, et al. Long-term follow up of human T-cell responses to conserved HIV-1 regions elicited by DNA/simian adenovirus/MVA vaccine regimens. PLoS One 2017; 12:e0181382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sette A, Crotty S. Immunological memory to SARS-CoV-2 infection and COVID-19 vaccines. Immunol Rev 2022; 310:27–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Guerrera G, Picozza M, D’Orso S, et al. BNT162b2 vaccination induces durable SARS-CoV-2-specific T cells with a stem cell memory phenotype. Sci Immunol 2021; 6:eabl5344. [DOI] [PubMed] [Google Scholar]
- 117▪.Reinscheid M, Luxenburger H, Karl V, et al. COVID-19 mRNA booster vaccine induces transient CD8+ T effector cell responses while conserving the memory pool for subsequent reactivation. Nat Commun 2022; 13:4631. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study characterizes the CD8+ T cell response generated by third and fourth booster shots of the COVID-19 mRNA vaccine platform. These results demonstrate that Spike-specific CD8+ T cells elicited by mRNA vaccination form a stable memory population that is rapidly recalled and cross-reactive to variants of concern during breakthrough infection.
- 118▪▪.Koutsakos M, Reynaldi A, Lee WS, et al. SARS-CoV-2 breakthrough infection induces rapid memory and de novo T cell responses. Immunity 2023; 56:879–892. e4. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study characterizes CD8+ T cell responses following breakthrough infection with Delta and Omicron variants, demonstrating that LNP/mRNA vaccines can generate memory CD8+ T cells that rapidly respond to infection with viral variants that evade antibody responses.
- 119.Oberhardt V, Luxenburger H, Kemming J, et al. Rapid and stable mobilization of CD8+ T cells by SARS-CoV-2 mRNA vaccine. Nature 2021; 597:268–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lorentzen CL, Haanen JB, Met Ö, Svane IM. Clinical advances and ongoing trials of mRNA vaccines for cancer treatment. Lancet Oncol 2022; 23:e450–e458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Chen J, Ye Z, Huang C, et al. Lipid nanoparticle-mediated lymph node-targeting delivery of mRNA cancer vaccine elicits robust CD8+ T cell response. Proc Natl Acad Sci U S A 2022; 119:e2207841119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Valentin A, Bergamaschi C, Rosati M, et al. Comparative immunogenicity of an mRNA/LNP and a DNA vaccine targeting HIV gag conserved elements in macaques. Front Immunol 2022; 13:945706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123▪▪.Künzli M, O’Flanagan SD, LaRue M, et al. Route of self-amplifying mRNA vaccination modulates the establishment of pulmonary resident memory CD8 and CD4 T cells. Sci Immunol 2022; 7:eadd3075. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that intramuscular vaccination with a self-amplifying mRNA vaccine can generate tissue resident memory that can be further expanded with intranasal administration, a concept relevant to HIV vaccine design.
- 124.Amaya L, Grigoryan L, Li Z, et al. Circular RNA vaccine induces potent T cell responses. Proc Natl Acad Sci U S A 2023; 120:e2302191120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Moyo N, Vogel AB, Buus S, et al. Efficient induction of T cells against conserved HIV-1 regions by mosaic vaccines delivered as self-amplifying mRNA. Mol Ther Methods Clin Dev 2019; 12:32–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Barouch DH, Tomaka FL, Wegmann F, et al. Evaluation of a mosaic HIV-1 vaccine in a multicentre, randomised, double-blind, placebo-controlled, phase 1/2a clinical trial (APPROACH) and in rhesus monkeys (NHP 13–19). Lancet Lond Engl 2018; 392:232–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Mutua G, Farah B, Langat R, et al. Broad HIV-1 inhibition in vitro by vaccine-elicited CD8(+) T cells in African adults. Mol Ther Methods Clin Dev 2016; 3:16061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Xu S, Carpenter MC, Spreng RL, et al. Impact of adjuvants on the biophysical and functional characteristics of HIV vaccine-elicited antibodies in humans. NPJ Vaccines 2022; 7:90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Bailón L, Llano A, Cedeño S, et al. Safety, immunogenicity and effect on viral rebound of HTI vaccines in early treated HIV-1 infection: a randomized, placebo-controlled phase 1 trial. Nat Med 2022; 28:2611–2621. [DOI] [PubMed] [Google Scholar]
- 130.Wee EG, Moyo NA, Saunders KO, et al. Parallel induction of CH505 B cell ontogeny-guided neutralizing antibodies and tHIVconsvX conserved mosaic-specific T cells against HIV-1. Mol Ther Methods Clin Dev 2019; 14:148–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Cohen KW, Tian Y, Thayer C, et al. Th2-biased transcriptional profile predicts HIV envelope-specific polyfunctional CD4+ T cells that correlated with reduced risk of infection in RV144 Trial. J Immunol Baltim Md 1950 2022; 209:526–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132▪▪.Kallas EG, Grunenberg NA, Yu C, et al. Antigenic competition in CD4+ T cell responses in a randomized, multicenter, double-blind clinical HIV vaccine trial. Sci Transl Med 2019; 11:eaaw1673. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study reports on results from the HVTN 084 trial, investigating whether inclusion of Env detracts from the magnitude and breadth of T cell responses to Gag/Pol. The authors found that inclusion of Env reduced breadth of T cell responses and specifically reduced response rate, magnitude, and cytokine expression in CD4+ T cells.
- 133. Chew KW, Reuschel E, Purwar M, et al. Including Env in an HIV therapeutic vaccine blunts Gag/Pol-specific T cell responses [Internet]. Conf Retroviruses Opportun Infect 2022; Virtual. https://www.croiconference.org/abstract/including-env-in-an-hiv-therapeutic-vaccine-blunts-gag-pol-specific-t-cell-responses/ [Google Scholar]
- 134.Churchyard GJ, Morgan C, Adams E, et al. A phase IIA randomized clinical trial of a multiclade HIV-1 DNA prime followed by a multiclade rAd5 HIV-1 vaccine boost in healthy adults (HVTN204). PLoS One 2011; 6:e21225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Casazza JP, Bowman KA, Adzaku S, et al. Therapeutic vaccination expands and improves the function of the HIV-specific memory T-cell repertoire. J Infect Dis 2013; 207:1829–1840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kalams SA, Parker SD, Elizaga M, et al. Safety and comparative immunogenicity of an HIV-1 DNA vaccine in combination with plasmid interleukin 12 and impact of intramuscular electroporation for delivery. J Infect Dis 2013; 208:818–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hancock G, Morón-López S, Kopycinski J, et al. Evaluation of the immunogenicity and impact on the latent HIV-1 reservoir of a conserved region vaccine, MVA.HIVconsv, in antiretroviral therapy-treated subjects. J Int AIDS Soc 2017; 20:21171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Mothe B, Manzardo C, Sanchez-Bernabeu A, et al. Therapeutic vaccination refocuses T-cell responses towards conserved regions of HIV-1 in early treated individuals (BCN 01 study). EClinicalMedicine 2019; 11:65–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mothe B, Rosás-Umbert M, Coll P, et al. HIVconsv vaccines and romidepsin in early-treated HIV-1-infected individuals: safety, immunogenicity and effect on the viral reservoir (Study BCN02). Front Immunol 2020; 11:823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Fidler S, Stöhr W, Pace M, et al. Antiretroviral therapy alone versus antiretroviral therapy with a kick and kill approach, on measures of the HIV reservoir in participants with recent HIV infection (the RIVER trial): a phase 2, randomised trial. Lancet Lond Engl 2020; 395:888–898. [DOI] [PubMed] [Google Scholar]
- 141.Jacobson JM, Zheng L, Wilson CC, et al. The safety and immunogenicity of an interleukin-12-enhanced multiantigen DNA vaccine delivered by electroporation for the treatment of HIV-1 infection. J Acquir Immune Defic Syndr 19992016; 71:163–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Edupuganti S, C De Rosa S, Elizaga M, et al. Intramuscular and intradermal electroporation of HIV-1 PENNVAX-GP® DNA vaccine and IL-12 is safe, tolerable, acceptable in healthy adults. Vaccines 2020; 8:741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Colby DJ, Sarnecki M, Barouch DH, et al. Safety and immunogenicity of Ad26 and MVA vaccines in acutely treated HIV and effect on viral rebound after antiretroviral therapy interruption. Nat Med 2020; 26:498–501. [DOI] [PubMed] [Google Scholar]
- 144.Campion SL, Brenna E, Thomson E, et al. Preexisting memory CD4+ T cells contribute to the primary response in an HIV-1 vaccine trial. J Clin Invest 2021; 131:e150823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145▪.Stieh DJ, Barouch DH, Comeaux C, et al. Safety and immunogenicity of Ad26-vectored HIV vaccine with mosaic immunogens and a novel mosaic envelope protein in HIV-uninfected adults: a phase 1/2a study. J Infect Dis 2023; 227:939–950. [DOI] [PMC free article] [PubMed] [Google Scholar]; Results from the ASCENT trial (HVTN 118), a predecessor of the Mosaico trial, focused on broadening B and T cell responses to Env using a mosaic Env protein boost.
- 146.Wilson GJ, Rodriguez B, Li SS, et al. Cellular and humoral responses to an HIV DNA prime by electroporation boosted with recombinant vesicular stomatitis virus expressing HIV subtype C Env in a randomized controlled clinical trial. Vaccine 2023; 41:2696–2706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Xu Y, Samir S, Weideman AMK, et al. Conserved-region MVA vaccines can shift HIV T cell immunodominance in PWH on ART - the M&M Study. J Immunol 2022; 208: (Suppl 1): 64.15–64.15. [Google Scholar]
- 148. Mothe Pujades B, Curran A, López JC, et al. A placebo-controlled randomized trial of the HTI immunogen vaccine and VESATOLIMOD [Internet]. Conf Retroviruses Opportun Infect 2023; 19 February 2023; Seattle, Washington. https://www.croiconference.org/abstract/a-placebo-controlled-randomized-trial-of-the-hti-immunogen-vaccine-and-vesatolimod/ [Google Scholar]
- 149.Hu X, Valentin A, Cai Y, et al. DNA vaccine-induced long-lasting cytotoxic T cells targeting conserved elements of human immunodeficiency virus Gag are boosted upon DNA or recombinant modified Vaccinia Ankara vaccination. Hum Gene Ther 2018; 29:1029–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]


