ADA-1 enhances HIV specific IgG and Antibody Dependent Complement Deposition (ADCD)
To evaluate the effect of ADA-1 on HIV specific humoral responses, 6–8-week-old female BALB/c mice were immunized thrice with 1µg pBG505-TTT or 5 µg pBG505-TTT alone or in combination with 10 µg pADA. Control mice were immunized with 15 µg Pvax, an empty plasmid vector, to ensure mice received equal total amounts of DNA and to control for any nonspecific DNA-induced innate immune responses. Mice were bled on day 14 post 1 immunization (D14P1), day 21 post 2 immunizations (D21P2), and sacrificed on day 14 post 3 immunizations (D14P3) to evaluate SOSIP-specific antibody magnitude, durability, and function via ELISA, ADCD (Fig. 1a) or neutralization assays (Supplemental Fig. 1d). Consistency between ELISA assays was confirmed using the human monoclonal antibody VRC01 as a positive control for assay validation (Supplemental Fig. 1a-b).
Mice co-immunized with pADA and 1 µg pBG505-TTT exhibited significantly increased SOSIP-specific IgG (mean O.D.=1.482) compared to mice immunized with 1 µg pBG505-TTT alone (mean O.D.=0.184) at both D21P2 (**p = 0.0022) and D14P3 (*p = 0.0289). However, mice co-immunized with pADA and 5 µg pBG505-TTT exhibited comparable levels of SOSIP-specific IgG (mean O.D.=1.063) to mice immunized with 5 µg pBG505-TTT alone (mean O.D.=0.998) suggesting a plateau in the humoral response with 5 µg pBG505-TTT. The ability of pADA to significantly enhance humoral responses with 1 µg pBG505-TTT (mean O.D.=1.482) to levels comparable with 5 µg pBG505-TTT (mean O.D.=0.998) clearly demonstrates a dose sparing effect with pADA (Fig. 1b). To evaluate the durability of the SOSIP-specific IgG induced following immunization, an area under the curve (AUC) analysis was performed on SOSIP-specific IgG that was measured across all timepoints (D14P1, D21P2, and D14P3). Mice co-immunized with pADA and 1 µg pBG505-TTT (mean AUC = 65.47) exhibited significantly increased SOSIP-specific IgG over time (**p = 0.0027) compared to mice immunized with 1 µg pBG505-TTT alone (mean AUC = 10.36) suggesting that ADA-1 induces more durable humoral responses (Fig. 1c).
To evaluate the functionality of SOSIP-specific Ab, we performed an ADCD assay using sera from immunized mice at D21P2 and D14P3. At the D21P2 timepoint, mice co-immunized with pADA and 5 µg pBG505-TTT produced SOSIP-specific Ab with the most capacity to induce ADCD (mean MFI = 331.14) with a trend toward increased ADCD compared to mice that received 5 µg pBG505-TTT alone (mean MFI = 232.38), although not significant. Mice co-immunized with 1 µg pBG505-TTT and pADA (mean MFI = 201.48) did not exhibit increased ADCD compared to mice immunized with 1 µg pBG505-TTT alone (mean MFI = 243.39). Compared to pVax immunized mice, mice immunized with 1µg pBG505-TTT (*p = 0.0307), 5 µg pBG505-TTT (**p = 0.0038) and 5 µg pBG505-TTT with pADA (***p = 0.0001) had significantly increased ADCD. This suggests the ability of ADA-1 to modestly increase ADCD particularly when co-administered with 5 µg pBG505-TTT. At the D14P3 timepoint, ADCD capacity contracts across all immunization groups compared to the D21P2 timepoint and there is no difference between mice immunized with pBG505-TTT alone or co-immunized with pADA (Fig. 1d).
In addition to ADCD, Ab neutralization capacity was evaluated. Mice immunized 5 µg pBG505-TTT alone (***p = 0.0006, mean ID50 = 156.29) and with pADA (**p = 0.0014, mean ID50 = 127.59) had significantly increased neutralization against the tier 1A pseudovirus Q461e2TAIV compared to Pvax mice. However, there was no difference in neutralization capacity between mice that received pBG505 alone or with pADA (Supplemental Fig. 1d). Additionally, we observed no correlation between SOSIP-specific IgG magnitude and neutralization (Supplemental Fig. 1e). Although this study strictly used female BALBc mice, previous studies using pADA in male and female mice did not show any sex biases nor any species (C57BL/6 or BALBc) biases in humoral or cellular responses [13]. Together these data demonstrate that pADA is dose sparing, enhances SOSIP-specific IgG magnitude and durability as well as modestly increases the ability of SOSIP-specific antibody to induce ADCD.
ADA-1 enhances HIV specific antibody isotype switching
To evaluate if ADA-1 enhances isotype switching, we measured SOSIP-specific IgG1, IgG2a, IgG2b and IgG3 in the sera of immunized mice as shown in Fig. 1a. Mice co-immunized with pADA and 1 µg pBG505-TTT exhibited significantly increased SOSIP-specific IgG1 at D21P2 (*p = 0.0137, mean O.D.=1.186) and a near significant increase (p = 0.0939, mean O.D.=1.057) at D14P3 compared to mice immunized with 1 µg pBG505-TTT alone (mean O.D. D21P2 = 0.104, mean O.D. D14P3 = 0.348). Similar to total SOSIP-specific IgG, SOSIP-specific IgG1 levels plateau at 5 µg pBG505-TTT (mean O.D. D21P2 = 1.341, mean O.D. D14P3 = 1.821) and we observe no further enhancement with pADA (mean O.D. D21P2 = 1.208, mean O.D. D14P3 = 2.059) (Fig. 2a).
At D21P2 we observed little to no SOSIP-specific IgG2a in both pADA receiving (mean O.D. 1 µg pBG505-TTT + pADA = 0.230, mean O.D. 5 µg pBG505-TTT + pADA = 0.255) and non-receiving counterparts (mean O.D. 1 µg pBG505-TTT = 0.113, mean O.D. 5 µg pBG505-TTT = 0.269). However, at D14P3 we see that only mice that were co-immunized with pADA develop SOSIP-specific IgG2a (mean O.D. 1 µg pBG505-TTT + pADA = 0.594, mean O.D. 5 µg pBG505-TTT + pADA = 0.493). The increase in SOSIP-specific IgG2a in pADA co-immunized mice was significant when compared to mice receiving 1 µg pBG505-TTT alone (*p = 0.0192) and near significant when compared to mice receiving 5 µg pBG505-TTT alone (p = 0.0513). These data demonstrate the ability of ADA-1 to uniquely induce SOSIP-specific IgG2a which is not achieved with pBG505-TTT alone (Fig. 2b). When correlating SOSIP-specific IgG1 to IgG2a production, we observe a significant positive correlation (**p = 0.008) (Fig. 2f).
Mice co-immunized with pADA and 1 µg pBG505-TTT exhibited significantly increased SOSIP-specific IgG2b at D21P2 (*p = 0.0101, mean O.D.=0.933) and a trend of increased SOSIP IgG2b (mean O.D.=1.121) at D14P3 compared to mice immunized with 1 µg pBG505-TTT alone (mean O.D. D21P2 = 0.142, mean O.D. D14P3 = 0.221). Similar to SOSIP-specific IgG and IgG1 responses, SOSIP-specific IgG2b levels plateau at 5 µg pBG505-TTT (mean O.D. D21P2 = 0.360, mean O.D. D14P3 = 0.608) and we observe no further enhancement with pADA (mean O.D. D21P2 = 0.332, mean O.D. D14P3 = 0.900) (Fig. 2c). When correlating SOSIP-specific IgG1 to and IgG2b production, we observe a significant positive correlation (**p = 0.001) between SOSIP-specific IgG1 and IgG2b (Fig. 2e). Regardless of timepoint or immunization, we observed minimal SOSIP-specific IgG3 production with no differences between mice immunized with pBG505-TTT alone or with pADA (Fig. 2d). Collectively, these data demonstrate that pADA enhances the isotype switching of SOSIP-specific Abs and uniquely drives IgG2a expression.
ADA-1 enhances HIV specific T cell responses
To quantify HIV-BG505 cell mediated immunity, we performed an IFNγ ELISpot assay using splenocytes from immunized mice as outlined in Fig. 1a. Spleens were harvested D14P3 and stimulated with peptide pools encompassing the HIV BG505 protein. HIV specific IFNγ production was quantified by spot forming units (SFUs) per million splenocytes using an ELISpot assay. In line with the humoral responses (Fig. 1–2), mice co-immunized with pADA and 1 µg pBG505-TTT had significantly increased IFNγ SFUs (*p = 0.0455, mean SFU = 1785) compared to mice with 1 µg pBG505-TTT alone (mean SFU = 958). Similar to humoral responses, we observed a plateau in SOSIP-specific IFNγ production in mice immunized with 5 µg pBG505-TTT alone (mean SFU = 2496) or with pADA (mean SFU = 2312) (Fig. 3a). These data suggest that ADA-1 co-delivery can enhance SOSIP-specific cellular IFNγ production and humoral responses (Fig. 1) in a dose sparing manner.
To understand immunogenic epitopes driving T cell responses, we colored the BG505 peptide pools onto the corresponding regions of the BG505 protein using a cryo-EM structure of a BG505 SOSIP.664 trimer (PDB ID: 6V0R). The peptides included in pool 5 have not been structurally resolved on the BG505 protein and are therefore not included. To determine the CD4 binding site, a cryo-EM structure of gp140 bound to the extracellular domains of CD4 (PDB ID: 7T0O) was aligned to the BG505 SOSIP.664 trimer (Fig. 3b-c). T cell responses against pBG505-TTT were primarily driven by the BG505 peptide pool 2 and pool 3. Pool 2 and 3 spanned the majority of the gp120 subunit. Notably, these peptide pools spanned important epitopes of BG505 including variable loops (V1/2, V3, V4 and V5), conserved domains (C2, C3, C4 and C5) and the CD4 binding site (Fig. 3b-c). These responses against peptide pool 2 and pool 3 were not necessarily enhanced with ADA-1, however, these findings demonstrate the ability of pBG505-TTT to drive T cell responses against important epitopes of the BG505 protein. Studies performing T cell epitope mapping of BG505 have been limited but have demonstrated major T cell epitopes within BG505 as C1, C2, C4, C5, V4 in gp120 and HR1 in gp41[42] which is in line with our findings presented here.
ADA-1 enhances HIV specific T cell polyfunctionality and degranulation
To further quantify HIV-BG505 cell-mediated immunity, we performed intracellular cytokine staining (ICS) to determine monofunctionality (IFNγ, TNFα or IL-2 expression, Supplemental Fig. 3) and polyfunctionality (IFNγ and TNFα or IFNγ, TNFα and IL-2 expression) of both CD4+ (Fig. 4a-b) and CD8+ (Fig. 4c-d) T cells using splenocytes from immunized mice as outlined in Fig. 1A. Spleens were harvested D14P3 and stimulated with peptide pools encompassing the HIV BG505 protein. Simultaneously, cells were stained for CD62L and CD44 to quantify central memory (CD62L+, CD44+) and effector memory (CD62L−, CD44+) T cells (Supplemental Fig. 7). The gating strategy used is shown in Supplemental Fig. 6. Cells were also stained for CD107a, a degranulation marker, to evaluate CD8+ T cell cytotoxicity (Fig. 4e). The gating strategy used is displayed in Supplemental Fig. 2.
When evaluating monofunctionality of CD4+ T cells, ADA-1 co-immunization resulted in increased frequencies of CD4+ T cells producing IFNγ and TNFα. Mice co-immunized with pADA and 1µg pBG505-TTT has significantly increased frequencies of IFNγ+ CD4+ T cells compared to mice immunized with 1 µg pBG505-TTT (*p = 0.0351) or Pvax (*p = 0.0256) (Supplemental Fig. 3a). Mice co-immunized with pADA and 5 µg pBG505-TTT had significantly increased frequencies of TNFα+ CD4+ T cells compared to mice immunized with 5 µg pBG505-TTT (*p = 0.0499) or Pvax (**p = 0.0014) (Supplemental Fig. 3b). We observed a trend of increased IL2+ CD4+ T cell frequencies with pADA and 5 µg pBG505-TTT, although not significant when compared to mice receiving 5 µg pBG505-TTT alone (Supplemental Fig. 3c).
When evaluating polyfunctionality of CD4+ T cells, we found that mice co-immunized with pADA and 5 µg pBG505-TTT had significantly increased frequencies of IFNγ+ TNFα+ CD4+ T cells compared to mice that were immunized with pVax (**p = 0.0047) and 5 µg pBG505-TTT alone (**p = 0.0030) (Fig. 4a). Mice co-immunized with pADA and 1 µg pBG505-TTT had significantly increased frequencies of IFNγ+ TNFα+ CD4+ T cells compared to pVax (*p = 0.0120) and an increasing trend when compared to mice immunized with 1 µg pBG505-TTT alone although not significant (Fig. 4a). Mice co-immunized with pADA and 5 µg pBG505-TTT had significantly increased frequencies of IFNγ+ TNFα+ IL2+ CD4+ T cells compared to mice that were immunized with Pvax (**p = 0.0023) 5 µg pBG505-TTT alone (*p = 0.0389) (Fig. 4b).
In the CD8+ T cell compartment, we observed no differences in monofunctionality between mice that had been co-immunized with pADA and mice immunized with pBG505 (Supplemental Fig. 3d-f). In terms of CD8+ T cell polyfunctionality, we observed that mice co-immunized with pADA and 1 µg pBG505-TTT had significantly increased frequencies of IFNγ+ TNFα+ CD8+ T cell compared to mice immunized with Pvax (*p = 0.0430) and 1 µg pBG505-TTT alone (*p = 0.0430) (Fig. 4c). When evaluating CD8+ T cell producing IFNγ, TNFα, and IL-2, however, we observed no differences between mice co-immunized with pADA or immunized with pBG505 alone (Fig. 4d). Mice co-immunized with pADA and 5 µg pBG505-TTT had significantly increased frequencies of CD8+ T cells expressing the degranulation marker CD107a compared to mice immunized with Pvax (*p = 0.0258) and 5 µg pBG505-TTT alone (*p = 0.0420) (Fig. 4e). Enhanced CD107a expression with pADA co-immunization suggests that ADA-1 increases the ability of CD8+ T cell to degranulate and to elicit cytotoxicity [43].
When evaluating central and effector memory T cell responses, we observed no effect on CD4+ central memory (CD62L+, CD44+) T cells (Supplemental Fig. 7a) with or without pADA. Mice co-immunized with 1 µg pBG505-TTT (*p = 0.0115) or 5 µg pBG505-TTT (*p = 0.0263) and pADA did have significant increases in frequencies of CD4+ effector memory (CD62L−, CD44+) T cells compared to Pvax immunized mice. These increases with pADA however, were not significant when compared to mice immunized with 1 µg pBG505-TTT or 5 µg pBG505-TTT alone (Supplemental Fig. 7b). Similar to CD4+ T cell central memory, we observed no effect on CD8+ T cell central memory (CD62L+, CD44+) frequencies (Supplemental Fig. 7c) with or without pADA. In contrast to CD8+ T cell central memory, we observed a significant increase in frequencies of CD8+ effector memory (CD62L−, CD44+) T cells with pADA co-immunization at both the 1 µg pBG505-TTT (***p = 0.0002) and 5 µg pBG505-TTT (*p = 0.0289) doses compared to mice immunized with 1 µg pBG505-TTT or 5 µg pBG505-TTT alone (Supplemental Fig. 7d). Frequencies of CD8+ effector memory (CD62L−, CD44+) T cells were also significantly increased in mice immunized with 1 µg pBG505-TTT and pADA (***p = 0.0002) and 5 µg pBG505-TTT + pADA (**p = 0.0047) doses compared to mice immunized with Pvax.
Taken together, these data demonstrate that ADA-1 enhances HIV specific T cell cytokine production and polyfunctionality of effector T cells. These data also suggest that ADA-1 enhances CD4+ and CD8+ effector memory T cell responses but has little to no effect on central memory T cell responses.
ADA-1 enhances HIV specific T cell activation and memory B cell frequencies
To further quantify HIV-BG505 cell mediated immunity, we performed an activation induced marker (AIM) assay and quantified SOSIP-specific memory B cells using flow cytometry. The AIM assay, as previously reported [44], is used to assess antigen specific activation of CD4+ T cells through the upregulation of activation markers such as PDL1, OX40 and CD25 upon stimulation. This assay has been described to enrich and monitor antigen specific TFH cells. For these experiments, splenocytes from immunized mice as outlined in Fig. 1a were stimulated with peptide pools encompassing the HIV BG505 protein and PDL1, OX40 and CD25 expression on CD+ T cells was quantified via flow cytometry. The gating strategy used is shown in Supplemental Fig. 4.
Mice co-immunized with pADA and 5 µg pBG505-TTT had significantly increased frequencies of CD4+ T cells expressing PDL1 and OX40 (**p = 0.0022, mean = 0.401%) compared to mice immunized with 5 µg pBG505-TTT alone (mean = 0.217%)(Fig. 5a). Mice co-immunized with pADA and 5 µg pBG505-TTT also had significantly increased frequencies of CD4+ T cells expressing PDL1 and CD25 (*p = 0.0280, mean = 0.253%) (Fig. 5b) compared to mice immunized with 5 µg pBG505-TTT alone (mean = 0.146%).
To evaluate SOSIP-specific memory B cell responses, we stained splenocytes from immunized mice as outlined in Fig. 1a with fluorescently labeled multimerized probes specific to SOSIP and defined memory B cells as splenocytes that were CD19+IgD− via flow cytometry.
In line with the humoral response (Fig. 1–2), mice co-immunized with pADA and 1 µg pBG505-TTT had significantly increased frequencies of SOSIP-specific memory B cells (*p = 0.0382, mean = 0.022%) compared to mice immunized with 1 µg pBG505-TTT alone (mean = 0.007%). At the 5 µg pBG505-TTT dose, we observed no difference in SOSIP-specific memory B cell frequencies in mice co-immunized with pADA (mean = 0.025%) or immunized with 5 µg pBG505-TTT alone (mean = 0.032%) (Fig. 5c).
Taken together, these data demonstrate that ADA-1 enhances HIV-specific CD4+ T activation and SOSIP-specific memory B cells when antigen dose sparing is applied.
ADA-1 induces qualitative differences in the neutralization capacity of HIV-specific antibodies
We have previously demonstrated that co-immunization with a plasmid-encoded monomeric gp160 HIV envelope DNA vaccine, recombinant gp160 protein, and pADA in mice resulted in enhanced neutralization of the tier 1A pseudovirus MW965 [12]. In these studies, enhanced neutralization was only the case upon co-immunization with DNA, protein and pADA and was not the case with DNA alone, DNA and protein or DNA and pADA [12]. For this reason, we believed that to see any qualitative difference in Ab responses within a BALB/c mouse model, immunization with DNA, protein and an adjuvant would be necessary. To address this, we immunized mice simultaneously with 10 µg pBG505-TTT alone or with pADA in the right tibialis anterior (TA) muscle and 10 µg rBG505 with alum or with the MF59-like adjuvant AddaVax in the left TA muscle. To understand how pADA performs compared to Alum and AddaVax, we immunized a separate group of mice with 10 µg pBG505-TTT alone or with pADA and no protein co-immunization. As a negative control, we immunized mice with 20µg of Pvax. Mice were immunized thrice and bled on D21P2 and sacrificed at D14P3 (Fig. 6a).
When evaluating SOSIP-specific IgG, across all immunization groups we observed no difference between mice that received pADA or their non-pADA receiving counterparts (Fig. 6b). This is likely due to the higher dose of pBG505-TTT (10 µg) used for this experiment as we observed a plateau in humoral responses with 5 µg pBG505-TTT (Fig. 1–2). We also evaluated SOSIP-specific IgA responses and observed no difference between mice that received pADA or their non-pADA receiving counterparts. We did, however, observe a significant increase in SOSIP-specific IgA in mice immunized with pBG505-TTT and pADA (*p = 0.0307) and mice immunized with pBG505, rBG505, Addavax and pADA (*p = 0.0121) compared to Pvax immunized mice. There was an observed trend toward increased SOSIP-specific IgA in pADA co-immunized mice compared to their non-ADA-1 receiving counterparts although not significant (Supplemental Fig. 1c).
When evaluating Ab functionality by pseudoviral neutralization assay, ADA-1 did increase neutralization of the Tier 1A pseudovirus Q461eTAIV to a near significant level (p = 0.07) compared to mice immunized with pBG505-TTT, rBG505, and alum. Mice co-immunized with pBG505-TTT, rBG505, and AddaVax had robust neutralization that was not further improved with pADA (Fig. 6c). We observe no correlation between SOSIP-specific IgG and neutralization ID50 demonstrating that the increased Ab quality with pADA is not a direct result of increased Ab magnitude (Fig. 6d-f). We did not observe neutralization in any immunization groups against the Tier 1B virus Q23env17 and the autologous BG505 virus (Supplementary Table 1–2) which could in part be due to shortcomings of using wild-type mouse models to evaluate the breadth of antibody neutralization. Collectively these data demonstrate the ability of ADA-1 to enhance the quality of SOSIP-specific Abs by improving neutralization capacity particularly against Tier 1A pseudovirus Q461eTAIV.