Weekly Digests
‹ Back to September

Better together: Vaccination with linked CD4+ and CD8+ T cell neoantigens synergizes with anti-PD-1

September 20, 2023

Neoantigen vaccination and immune checkpoint blockade (ICB) are both strategies that can be employed to enhance antitumor T cell responses, but both are often met with resistance, and their use in combination has not been fully explored. Investigating these strategies in tumors with low mutation burdens, Dolina et al. found that immunization with neoantigens that are naturally recognized by both CD4+ and CD8+ T cells could induce antitumor immunity and overcome resistance to ICB. Interestingly, the CD4+ target antigen did not need to be tumor-specific, so long as it was linked to a tumor-specific CD8+ target antigen. The results of this study were recently published in The Journal of Clinical Investigation.

For their studies, Dolina et al. selected squamous cell carcinoma VII (SCC VII) – a spontaneously arising MHC-II- murine tumor that resembles human HNSCC and resists both chemo- and immunotherapy – as their tumor model. This model also contains tumor-initiating cancer stem cells (tCSC), which are marked by high CD44 and PD-L1 expression, are inherently resistant to treatments, and contribute to tumor heterogeneity. Immunizing mice with irradiated SCC VII prior to challenge with live tumor did not induce antitumor immunity, though adding polyI:C to the immunization led to rejection of the challenge, dependent on CD4+ and CD8+ T cells, suggesting that this model is naturally weakly immunogenic.

To detect neoantigens in SCC VII, the researchers used genomic sequencing and functional analysis. From the 39 mutations that were found to reach their expression threshold, the researchers generated 91 candidate peptides and tested them as targets for T cells by using them to restimulate mononuclear cells isolated from mice that had resisted a tumor challenge. Ultimately, the researchers were able to identify four mutated genes (including driver mutations) that produced five mutant peptides recognized by natural T cell responses. Immunization with a prime and boost containing a combination of these 5 peptides + polyI:C protected mice from challenge with SCC VII. Further, one of the peptides alone, MUT_48 + polyI:C was found to be sufficient to protect mice from tumor challenge.

Assessing the responses to these neoantigens, the researchers found that Mut_48 alone was recognized by both CD4+ and CD8+ T cells, while the others were recognized only by CD4+ T cells. Some CD8+ T cells also recognized two additional peptides, Mut_72 and Mut_73; however, these peptides were not capable of conferring protective immunity in vivo.

Digging deeper into the specific epitopes of MUT_48 that were recognized by CD4+ and CD8+ T cells, the researchers developed a panel of 10-mer and 15-mer peptides containing the MUT_48 H129Q mutation. CD8+ T cells identified the MUT_48.10 10-mer and CD8+ T cell minimal epitope (presented on H-2Kk). While CD4+ T cells did not recognize the MUT_48.10 10-mer, they did recognize the Mut_48.5 15-mer (presented on I-Ak), which contained the Mut_48.10 10-mer sequence. In vitro, the Mut_48.5 15-mer could elicit IFNγ responses from both CD4+ and CD8+ T cells. Similarly, in vivo, immunization with the Mut_48.5 15-mer was as protective against SCC VII as the full Mut_48 20-mer, while immunization with Mut_48.10 (recognized only by CD8+ T cells) was only partially protective, suggesting that CD4+ T cells were required for full prophylactic immunity.

To determine whether CD4+ T cell help needed to be tumor-specific, mice were immunized with the Mut_48.10 CD8+ T cell minimal epitope plus either SCC VII-derived Mut_44 (recognized by CD4+ T cells only) or with the PAN-DR epitope, PADRE(X), which is universally immunogenic to CD4+ T cells in Black6 mice. Both of these strategies were effective, eliciting a similar degree of protection as MUT_48 against SCC VII, suggesting that the CD4+ T cell epitope does not need to be tumor-specific. Further, covalent linkage of CD4+ and CD8+ T cell antigens enhanced protection compared to untethered peptides, suggesting that the enhanced protection from dually recognized peptides like MUT_48 likely stems from presentation of both the CD8+ target antigen and CD4+ helper antigen by the same APC, consistent with Th-mediated licensing of APCs to induce enhanced CD8+ T cell responses.

Next, Dolina et al. evaluated prophylactic neoantigen vaccination followed by ICB. When mice were treated prophylactically with the 5-peptide neoantigen vaccine alone, about half of mice experienced a late-phase relapse with SCC VII. However, with the addition of anti-PD-1 shortly after tumor implantation, these relapses could be prevented. Anti-PD-1 also expedited the elimination of tumors, increased memory T cell responses at day 42, and supported epitope spreading to Mut_72 and Mut_73 – targets that were not included in the vaccine. Similar results were not observed with anti-CTLA-4 delivered shortly after implantation.

When the combination of neoantigen vaccination and anti-PD-1 was tested against large, established tumors, they again showed synergy, inducing complete and durable tumor clearance that could not be achieved with either treatment alone. In this setting, combination treatment was found to increase CD8+ T cells among TIL and enhance Mut_48-specific memory T cell responses. Further, it appeared to inhibit metastasis to regional LNs.

Investigating the phenotypes of responding T cells in this setting, the researchers found that Mut_48 vaccination reduced naive and certain effector memory CD4+ and CD8+ T cell populations (in line with priming), while anti-PD-1 subtly expanded existing populations in TIL, including precursor/progenitor T cells, intermediate/exhausted cells, and an effector memory population of follicular T helper cells. Conventional CD4+ TILs were not cytotoxic, fitting with their role as helpers in this setting. Further, the addition of anti-PD-1 abrogated the expansion of terminally exhausted cells that was induced by vaccination alone, instead steering populations towards less differentiated states. Combination treatment also expanded a small population of precursor/progenitor T cells in the spleen and tumor-draining lymph nodes.

While CD4+ T cells were required for full efficacy in this model, CD8+ T cells were the critical effectors. Suspecting that CD4+ T cells provided Th-1-based help through CD40-mediated signaling in APCs, the researchers showed that an agonistic anti-CD40 cross-linking could replace CD4+ T cell help to support CD8+ T cell-mediated tumor eradication.

Finally, looking at tCSCs, Dolina et al. showed that while neither Mut_48 vaccination nor PD-1 blockade individually could significantly increase killing of SCC VII CD44lo tumor cells or CD44hi tCSC, combination treatment could effectively target both subsets, suggesting that this strategy can overcome resistance mechanisms.

Overall, these results show that vaccinations with neoantigens naturally recognized by the immune system are more effective when peptides simultaneously stimulate helper CD4+ and effector CD8+ T cells, likely via the same APC. Further, neoantigen vaccination can synergize with anti-PD-1 to induce both prophylactic and therapeutic antitumor immunity, which could be translationally relevant.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, first author Joseph Dolina answered our questions. 

What was the most surprising finding of this study for you?
When scanning peptide reactivity for individual CD4+ and CD8+ epitopes, we were surprised that full protection from tumor burden was solely elicited by a neoantigen that concurrently stimulated both T cell subsets. Neither CD4+ nor CD8+ T cell neoantigen vaccination alone was sufficient to cure mice of cancer, and this made sense! This is how CD4+ T cell help works… synchronized loading of MHC I and II epitopes on the surface of the same antigen-presenting cell. Further, helped tumor-specific CD8+ T cells were more capable of forming memory and resisting exhaustion.

What is the outlook?
This work has immediate impacts into vaccine design for clinical trials – establishing the need to include verified CD4+ T cell neoepitopes for supporting stable antitumor CD8+ T cell responses. However, more research is needed to understand how peptide affinity and the quality of CD4+ T cell help affects CD8+ T cell differentiation in the periphery and once inside the harsh tumor microenvironment. Function-based methods for altering tumor vaccination behavior, as outlined in this work, can also be used to train neoantigen prediction algorithms to better model how T cells respond to tumor neoantigens.

What was the coolest thing you’ve learned (about) recently outside of work?
I recently became a parent to Joseph the 5th (yes there is a lineage of us) during the writing of this manuscript. He has opened my world and touched my heart to levels I never could have expected.

References:

Dolina JS, Lee J, Brightman SE, McArdle S, Hall SM, Thota RR, Zavala KS, Lanka M, Ramamoorthy Premlal AL, Greenbaum JA, Cohen EEW, Peters B, Schoenberger SP. Linked CD4+/CD8+ T cell neoantigen vaccination overcomes immune checkpoint blockade resistance and enables tumor regression. J Clin Invest. 2023 Sep 1.

In the Spotlight...

Tumor dynamics in patients with solid tumors treated with pembrolizumab beyond disease progression

Topp et al. evaluated the efficacy of pembrolizumab in 799 patients who received treatment beyond progressive disease per RECIST v1.1 across six solid tumor types, and showed that a proportion of patients respond to therapy beyond progression. In patients treated beyond progression, a majority of patients (64.8%–76.5%) showed stable target lesion dynamics, while a subset of patients (8.9% and 24.4%) showed a 30% or greater reduction in the sum of lesion diameters. ICI-responsive tumor types, such as melanoma, showed the highest response rates, and patients with longer durations of treatment showed deeper responses.

Contributed by Shishir Pant

Combination therapy with dendritic cell loaded-exosomes supplemented with PD-1 inhibition at different time points have superior antitumor effect in hepatocellular carcinoma

Chang et al. showed that anti-PD-1 therapy increased the antitumor efficacy of DC-TEX by restoring exhausted tumor antigen-specific CD8+ T cells in an orthotopic HCC mouse model. DC-TEX treatment increased the number of CD8+ T cells, activation-associated cytokines, and PD‑1+ CD8+ T cells, peaking at 72 h post treatment. Anti-PD-1 increased the proliferation and cytokine production of PD‑1+ CD8+ T cells maximally at 72 h in vitro. The combination of DC-TEX and anti-PD-1 showed superior antitumor response to HCC in mice when PD-1 Ab was administered 72 h (vs 24, 120, and 168 h) after DC-TEX, suggesting that the timing of PD-1 Ab administration impacts the antitumor effect.

Contributed by Shishir Pant

Overcoming lung cancer immunotherapy resistance by combining non-toxic variants of IL-12 and IL-2

Focusing on resistance mechanisms to cytokine therapies, Horton et al. found that systemic mouse serum albumin (MSA)-fused IL-12 therapy in a lung tumor model failed to control lung tumor growth due to low IL-12R levels on tumor-reactive T cells in the lung, while successfully controlling growth of flank tumors. Addition of MSA-IL-2 increased IL-12 binding to tumor-reactive T cells, and the combination increased tumor-reactive CD8+ T cell effector differentiation, decreased Tregs, and increased survival, but was associated with significant dose-limiting toxicity. Toxicity was alleviated using a combination of engineered IL-2 and IL-12 variants shown to have reduced dose-limiting toxicity.

Contributed by Katherine Turner

Endogenous H3.3K27M derived peptide restricted to HLA-A∗02:01 is insufficient for immune-targeting in diffuse midline glioma

A novel high-affinity scFv antibody targeting the histone 3.3 K27M25-35 neoepitope in the context of HLA-A*02:01 recognized peptide-pulsed APCs, but not H3.3K27M-mutated cell lines derived from patients with diffuse intrinsic pontine glioma (DIPG). Wang and Pandey et al. found that even though H3.3 is undergoing proteasomal degradation, and non-mutated H3.3-derived peptides are presented on the cell surface, the H3.3K27M25-35 neoepitope in not endogenously processed and presented in HLA-A*02:01+ DIPG cell lines. Immunopeptidomics identified a novel potential HLA-A*03:01-restricted H3.3K27M26-36 neoepitope that is processed and presented.

Contributed by Ute Burkhardt

Alternative splicing of its 5' UTR limits CD20 mRNA translation and enables resistance to CD20-directed immunotherapies

Investigating CD20 antigen escape mechanisms, Ang et al. identified four mRNA isoforms (V1-4) of CD20 with unique 5’-UTRs. V1 and V3 were the most abundant, and while V3 correlated with CD20 protein expression, V1 did not, and was found to contain features that inhibited polysome recruitment and limited translation. V1 yielded sufficient CD20 to be recognized by CD20-targeted CAR T cells, but allowed for escape from monoclonal (rituximab) and bispecific (mosunetuzumab) antibody therapies. Splice-switching Morpholino oligomers or reconstituting CD20 KO cells with V3 recovered CD20 expression. Evidence of a V3–V1 shift was also identified in relapse after treatment with mosunetuzumab.

Contributed by Lauren Hitchings

The CD58-CD2 axis is co-regulated with PD-L1 via CMTM6 and shapes anti-tumor immunity

By analyzing patient melanomas biopsied before or on ICB, and patient-derived melanoma cells and autologous ex vivo-expanded TILs in co-cultures and in a humanized mouse model, Ho, Melms and Rogava et al. showed that cancer cell CD58 was required for specific T cell-mediated antitumor activity. Cancer cell CD58 loss reduced intratumoral T cell infiltration, expansion, and differentiation, upregulated PD-L1, and boosted immune evasion. CD58 stability and PD-L1 upregulation upon CD58 loss were co-regulated by their competition for CMTM6 binding, which stabilized cell surface proteins via endosomal recycling and reduced lysosomal degradation.

Contributed by Paula Hochman

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.