Weekly Digests
‹ Back to June

Three’s company: immune triads license CD8+ T cell-mediated antitumor immunity

June 26, 2024

CD4+ T cells are known to “help” CD8+ T cells by licensing their cytotoxic functions in tumors, but exactly how this help is given is not entirely understood. To better understand how CD4+ T cells contribute to CD8+ T cell responses, Espinosa-Carrasco et al. developed a tumor and adoptive cell therapy (ACT) model in which they could study the relationship between these cells, and found that CD4+ T cells formed critical triads with CD8+ T cells and APCs in order to license CD8+ T cell-mediated cytotoxicity and antitumor immunity. Their results were recently published in Cancer Cell.

To begin, Espinosa-Carrasco et al. engineered the B16F10 melanoma model to express (1) the SIINFEKL peptide from the OVA antigen for recognition by OT-I CD8+ T cells, (2) the glycoprotein epitope GP61–80 from LCMV for recognition by SMARTA TCR-transgenic CD4+ T cells, and (3) a fluorescent tag. The resulting B16-OG cells grew aggressively upon implantation into mice, forming large tumors within two weeks. Mimicking clinical protocols for ACT, the researchers pre-treated mice with lymphodepleting chemotherapy, then transferred activated OT-I cells, SMARTA cells, or both. While transfer of either cell type alone allowed for continued tumor progression, the combination of CD8+ and CD4+ T cells resulted in complete tumor elimination and long-term tumor-free survival in 100% of mice. This effect was lost when OT-I cells were combined with CD4+ T cells specific for an irrelevant antigen, suggesting that both types of responses must be tumor antigen-specific for antitumor efficacy. Similar results were observed in an engineered MCA205-OG tumor model.

Looking within tumors, the researchers found that 8-9 days post-transfer, OT-I cell infiltration was similar, regardless of whether they were transferred with SMARTA cells. However, OT-I TILs that had been transferred alone produced few effector cytokines and expressed higher levels of inhibitory receptors and Tox, while those transferred in tandem with SMARTA cells produced much more IFNγ and TNFα and showed little to no expression of inhibitory receptors or Tox, suggesting that CD4+ T cells mediate a functional difference in CD8+ T cells. Further, this effect was observed in tumors, but not tdLNs, indicating that CD4+ T cell support is provided on-site in the TME.

To determine whether CD4+ T cells could reverse CD8+ T cell exhaustion, the researchers transferred OT-I cells into tumor-bearing mice, followed by transfer of SMARTA cells 10 days later. While tumors continued to progress with OT-I cells alone, the late addition of SMARTA cells rescued OT-I production of IFNγ and TNFα, and induced tumor regression. To better understand how this licensing occurred, the researchers performed RNAseq and found that the inclusion of SMARTA cells reduced the expression of various dysfunction-related transcription factors and inhibitory receptors, and instead induced expression of transcription factors and molecules associated with stem-like progenitor states. These results were also reflected in ATACseq data, which showed differentially accessible regions in exhaustion and stem/progenitor genes.

Evaluating the clinical relevance of these findings, the researchers compared profiles identified in mouse models with data from patients with metastatic melanoma lesions who received lymphodepleting chemotherapy followed by ACT. Here, the team identified a subset of CD39-CD69- stem-like TILs resembling OT-I cells that had been transferred with SMARTA cells. This subset in patients was associated with complete regression in responders, and was notably lacking in non-responders. In contrast, a CD39+CD69+ subset identified in non-responders resembled OT-I cells that were transferred without SMARTA cells in mice.

Next, the researchers investigated whether CD4+ T cells might be acting directly on tumor cells, but found that B16-OG tumor cells engineered to be MHC-II-deficient were eliminated with similar efficacy. However, when CD11c+ DCs were conditionally depleted in vivo, the antitumor effect of OT-I + SMARTA cells was lost, suggesting that APCs have a role in CD8+ T cell licensing. To separate out the presentation of the different tumor antigens by APCs, the researchers generated B16-O and B16-G tumor cell lines, each expressing only one of the respective model antigens, and then co-transferred both tumor cell lines into mice to generate heterogeneous B16 O+G tumors. Unlike the homogeneous B16-OG tumors B16 O+G tumors failed to respond to treatment with a combination of OT-I and SMARTA cells, despite similar antigen expression levels and similar tumor infiltration. In the B16 O+G tumors model, the OT-I cells co-transferred with SMARTA cells more closely resembled OT-I cells that had been transferred alone in the B16-OG model, expressing an exhausted/dysfunctional phenotype.

Confocal microscopic analysis of established B16 O+G tumors showed that there was little overlap between tumor regions expressing OVA versus GP antigens, suggesting that antigen-presenting cells in a given region would likely only pick up one antigen at a time, rather than both, as they would in the B16-OG model. Investigating whether co-presentation of antigens was the key to antitumor efficacy in the B16-OG model, the researchers developed a spatial analysis model in which cancer cells, OT-I cells, SMARTA cells, and APCs were each fluorescently tagged with unique markers. This revealed that in the B16-OG model, 3.5-fold more three-cell clusters, or “triads” of OT-I cells, SMARTA cells, and APCs formed than in the B16 O+G model. Further, when B16-OG cancer cells were injected into one flank while B16-O cancer cells were injected the opposite flank of the same mouse, only the B16-OG were eliminated by OT-I and SMARTA combination therapy, highlighting the importance of co-expression of tumor antigens on the same APCs within tumors to support CD8+ T cell-mediated antitumor immunity. Triad formation was also found to be relevant in responses to checkpoint blockade, as B16-OG, but not B16 O+G tumors were rejected upon treatment with anti-PD-1/anti-PD-L1.

Again turning to patient data, the researchers assessed the spatial orientation of CD4+ T cells (non-Tregs), CD8+ T cells, and APCs in patients with malignant plural mesothelioma who were treated with checkpoint blockade. While dyads of CD4+ T cells and APCs were not associated with responses, triad formation could delineate responders from non-responders. Further, PD-1+CD8+ T cells that were part of triads expressed higher levels of IFNγ and granzyme B compared to those not in triads.

Overall, these results suggest that the formation of triads of CD4+ T cells, CD8+ T cells, and APCs, in which APCs co-presenti relevant tumor antigens to each T cell type, allows for CD4+ T cell-mediated licensing and reprogramming of CD8+ T cells towards a more progenitor-like state, preventing and even reversing exhaustion and dysfunction. The formation of these triads was further associated with antitumor responses and clinical outcomes in mice and patients. Triad formation could therefore serve as both a biomarker for response and a target for improving various forms of cancer immunotherapy.

Write-up and image by Lauren Hitchings

References:

Espinosa-Carrasco G, Chiu E, Scrivo A, Zumbo P, Dave A, Betel D, Kang SW, Jang HJ, Hellmann MD, Burt BM, Lee HS, Schietinger A. Intratumoral immune triads are required for immunotherapy-mediated elimination of solid tumors. Cancer Cell. 2024 Jun 19.

In the Spotlight...

Botensilimab plus balstilimab in relapsed/refractory microsatellite stable metastatic colorectal cancer: a phase 1 trial

An Fc-enhanced anti-CTLA4 (botensilimab) designed to engage activating FcγRs of NK cells and APCs was combined with anti-PD-1 (balstilimab) in 148 patients with treatment-refractory MSS mCRC. Serious (grade 3+) TRAEs occurred in 35% of patients; the most common immune-related TRAEs (diarrhea and colitis) were well managed with infliximab (anti-TNF). The ORR was 17%, 45% of patients experienced SD, and the mPFS was 3.5 months. Active liver metastasis (LM) was a significant predictor of non-response. While the ORR was 0% and the mPFS was 1.4 months in patients with LM , the ORR in patients without LM was 22%, with an mPFS of 4.1 months.

Contributed by Morgan Janes

Immunological synapse formation between T regulatory cells and cancer-associated fibroblasts promotes tumour development

Varveri et al. investigated mechanisms used by CAFs to modulate antitumor immunity and contribute to cancer immunotherapy resistance. In multiple tumor models and patient biopsies, Foxp3+ Tregs preferentially accumulated in the TME in α-SMA+ CAF-rich areas, and formed synapses with α-SMA+ CAFs in an antigen-specific manner. Mechanistically, α-SMA+CAFs phagocytosed, processed, and presented tumor antigens via autophagy, which promoted Treg activation and proliferation. Ablation of CAF autophagy decreased Tregs and reprogrammed CAFs into a proinflammatory phenotype that potentiated the efficacy of dual ICB.

Contributed by Katherine Turner

Neoadjuvant Nivolumab and Ipilimumab in Resectable Stage III Melanoma

Blank and Lucas et al. report that two cycles of neoadju­vant ipilimumab plus nivolumab followed by therapeutic lymph node dissection and response-driven adjuvant nivolumab resulted in longer event-free survival (EFS) than the standard adju­vant nivolumab among patients with resectable, macroscopic, stage III melanoma. At a median follow-up of 9.9 months, the estimated 12-month EFS was 83.7% in the neoadjuvant group and 57.2% in the adjuvant group. Grade 3 or higher adverse events related to systemic treatment occurred in 29.7% of patients in the neoadjuvant group and 14.7% in the adjuvant group.

Contributed by Shishir Pant

Gene and protein sequence features augment HLA class I ligand predictions

Aiming to improve the predictability of HLA peptide ligands, Bresser et al. began with immunopeptidomics data from three melanoma cell lines, and investigated ligand-extrinsic hard-coded features of the encoding mRNA, including sequence composition/codon usage, translation-impacting RNA modifications (e.g., methylation), and contextual protein degradation signals (ubiquitination). Inclusion of these features produced a series of predictors that outperformed conventional binding affinity, equivalent to inclusion of “wet lab” RNA expression and translation data. The results were validated with external datasets.

Contributed by Ed Fritsch

Selective depletion of tumor-infiltrating regulatory T cells with BAY 3375968, a novel Fc-optimized anti-CCR8 antibody

Roider et al. demonstrated that CCR8 is predominantly expressed on activated tumor‑infiltrating regulatory T cells, and developed BAY 3375968, a novel fully human afucosylated monoclonal IgG1 anti-CCR8 antibody that effectively depleted Tregs. The preclinical antitumor activity of anti-mouse CCR8 antibodies was dependent on Fc-mediated effector functions, inducing antibody-dependent cellular cytotoxicity and phagocytosis. Combination of CCR8+ Treg depletion with immune checkpoint blockade enhanced antitumor responses, and induced long‑term and transplantable antitumor immunological memory.

Contributed by Shishir Pant

Neoantigen-specific stimulation of tumor-infiltrating lymphocytes enables effective TCR isolation and expansion while preserving stem-like memory phenotypes

Levin and Kim et al. developed a method for selective expansion of neoantigen-reactive CD4+ and CD8+ TILs with a broadened clonal repertoire, less exhaustion, and stem-like memory phenotypes by in vitro stimulation of bulk TILs with IL-2, IL-21, and APCs presenting selected p53 and RAS neoepitopes (“NeoExpand”). In contrast to TILs generated by conventional rapid expansion with OKT3, which reduced the frequencies of neoantigen-reactive T cells, NeoExpand TILs demonstrated improved antitumor efficacy in xenograft mouse models. Across 25 patients, NeoExpand facilitated the sensitive identification of rare and novel neoantigen-specific TCRs.

Contributed by Ute Burkhardt

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.