Weekly Digests
‹ Back to June

Strong and steady: robust TCR stimulation supports Tpex cell formation and maintenance

June 12, 2024

Progenitor exhausted T cells (Tpex), marked by high expression of Tcf1, Tox, and Ly108, help to replenish and maintain effector T cells in tumors, allowing for strong and sustained antitumor functions, including in the context of immunotherapy responses. However, exactly how these cells acquire and maintain this functional phenotype, and whether TCR engagement plays a role is not fully understood. To better understand Tpex cells, Lan et al. developed Lewis lung carcinoma (LLC) models expressing versions of the gp33 antigen that either optimally (M9) or suboptimally (C6M9) engaged the TCRs of gp33-specific T cells (P14). The results of their investigation were recently published in Nature Immunology.

To begin, Lan et al. implanted tumors expressing either M9 or C6M9 into wild-type and Rag-/- mice, followed by adoptive transfer of P14 cells 6 days later. In wild-type mice without P14 cells, the different tumors showed comparable growth, but in Rag-/- mice adoptively transferred with P14 cells, the M9-expressing tumors were better controlled. In wild-type mice with M9 tumors treated with P14 cells, P14 cells in tumor-draining lymph nodes (tdLNs) were more abundant, more proliferative, and expressed higher levels of activation markers CD44, PD-1, and Nr4a compared to in mice with C6M9 tumors. While Tpex cells were induced in both models, they became more enriched through day 12 in the M9 model, while they peaked at day 4 and then reduced in the C6M9 model, suggesting that robust TCR signaling preserved the Tpex phenotype in tdLNs.

Looking at the transcriptomic level, the researchers performed scRNAseq on P14s from the tdLN of tumor-bearing taken 8 days after transfer. Cells from M9-tdLN largely fell into clusters where Tpex gene signatures were highly enriched, while cells from C6M9-tdLN were slightly more proliferative, and fell more into central memory or acutely stimulated clusters, including a cluster that resembled Tex-KLR cells, found in chronic viral infections. Terminally differentiated cells were largely absent in tdLNs from either model.

Next looking at tumors, Lan et al. found that P14 cells more readily infiltrated M9-expressing tumors compared to C6M9-expressing tumors, and were more abundant in early tumors, though the discrepancy decreased with time. Cells in both tumors adopted exhaustion signatures; however, a higher proportion of Tpex cells was maintained in M9-expressing tumors, while a higher proportion of effector-like exhausted cells was established in C6M9-expressing tumors earlier on. Tpex showed higher Nr4a expression, indicative of stronger TCR signaling than exhausted T cells, suggesting that strong TCR signaling may help to maintain the Tpex phenotype in tumors.

Blockade of lymphocyte egress from tdLNs using FTY720 showed that Tpex frequency was reduced, but not lost in M9 tumors, with minimal effects in C6M9 tumors, suggesting that Tpex are both replenished from tdLNs and maintained in the TME. Supporting this, the researchers observed that when P14 cells that had been optimally primed in M9-tDLNs (about 90% Tpex) were adoptively transferred into mice bearing C6M9 tumors, they mostly lost the Tpex phenotype, instead transitioning into more terminal differentiation states with higher effector functions.

Using immunofluorescence imaging to interrogate the tumor immune landscape further, Lan et al. noted that a higher proportion of P14s in M9-expressing tumors colocalized with DCs, and became more activated when they were located adjacent to DC and B cell zones. This effect was not identified in C6M9-expressing tumors, suggesting that sustained optimal TCR engagement may be mediated through enhanced antigen-presentation by APCs.

Digging deeper into how TCR stimulation supports Tpex development and maintenance within cells, the researchers performed ATACseq to evaluate chromatin remodeling. At day 8, the epigenetic landscapes between antigen-experienced P14s from tdLNs in M9 and C6M9 models were distinct, as were the landscapes between antigen-experienced P14s from tdLNs and tumors from the same mice, suggesting that epigenetic programming is affected both by the quality of TCR engagement during priming, and by the tumor environment later on.

Looking at reprogramming related to priming in tdLNs, the researchers found that P14s in M9-tdLNs acquired more differentially expressed regions (DARs) and more DARs with higher fold changes. In Gene ontology enrichment analysis using the top 500 M9-tdLN-DARs, Tpex-associated genes were more accessible, while genes consistent with memory and quiescence were more accessible in P14s from C6M9-tdLNs. Further, M9-tdLN-DARs were enriched for the transcription factor binding motifs of Egr2 (Tex maintenance), and transcription factors that control activation-, effector-, and memory- associated genes. C6M9-tdLN-DARs, on the other hand, were enriched for motifs of transcription factors that repress CD8+ effector T cell functions.

Investigating whether TCR stimulation-induced chromatin remodeling was due to or accompanied by DNA methylation, the researchers performed whole-genome bisulfite sequencing (WGBS). Mirroring the chromatin remodeling data, methylation patterns were distinct between models and tissues, with more differentiated P14 cells from tumors undergoing progressive methylation reprogramming over time. In M9-tdLN, optimally primed P14 cells gained a higher number of demethylated regions (compared to P14s in C6M9-tdLN); were enriched for pathways involved in T cell activation, differentiation, and phospholipid transportation; and were hypomethylated at Tpex genes, corresponding with enhanced chromatin accessibility. Differentially methylated regions were also enriched for sites for AP1 family members and Egr1/Egr2 binding. Despite all this, the majority of differentially accessible regions (DARs; 87.6%) associated with differential TCR signal strength could not be linked to a significant change in DNA methylation, suggesting that histone modification is the predominant mechanism behind the chromatin remodeling.

While the differences in chromatin accessibility in P14 cells in tdLNs between M9 and C6M9 models were more pronounced, the differences in P14s infiltrating the tumors themselves were less distinct. Analysis of DARs suggested that Tpex cells from M9-expressing tumors were less differentiated, and showed little overlap with DARs from tdLNs. Enriched TF motifs were also distinct between models and tissues. Interestingly, Tcf1 was enriched in both the tumors and tdLNs of mice bearing M9-expressing tumors, and the openness of its binding sites were largely dependent upon TCR stimulation. Using CRISPR–Cas9 to disrupt the Tcf1 motifs in a region of Slamf6 (encoding Ly108) induced by TCR-stimulation, Ly108 expression and effector T cell differentiation upon adoptive transfer and tumor infiltration were abrogated, validating the functional relevance of Tpex-specific Tcf1 binding in maintaining tumor-reactive Tpex.

Overall, these results show that optimal TCR engagement plays a role in epigenetic imprinting that supports both Tpex formation during priming and Tpex maintenance and self-renewal in tumors. The effect in tumors was associated with proximity to DCs. Further, suboptimal TCR engagement could inhibit optimally primed Tpex cells, suggesting that maintaining optimal TCR engagement in tumors may be critical to antitumor efficacy.

Write-up and image by Lauren Hitchings

References:

Lan X, Mi T, Alli S, Guy C, Djekidel MN, Liu X, Boi S, Chowdhury P, He M, Zehn D, Feng Y, Youngblood B. Antitumor progenitor exhausted CD8+ T cells are sustained by TCR engagement. Nat Immunol. 2024 Jun;25.

In the Spotlight...

Engineering allorejection-resistant CAR-NKT cells from hematopoietic stem cells for off-the-shelf cancer immunotherapy

In a six-week, feeder-free process, human cord blood CD34+ HSCs were differentiated to NKT cells and engineered to express iNKT TCRs, anti-BCMA CARs, and IL-15, with knockout of B2M and CIITA to avoid HLA-based allo-rejection. Compared to standard CAR T cells, the CAR NKT cells avoided GvHD and allo-rejection by donor-mismatched PBMCs in mouse models, improving persistence and long-term multiple myeloma tumor control. Unlike CAR T cells, CAR NKT cells killed tumor cells through multiple targets: BCMA (via CAR), CD1d (via the invariant TCR, which also depleted CD1d-expressing TAMs and MDSCs), and NK receptor ligands.

Contributed by Alex Najibi

Glycoengineering-based anti-PD-1-iRGD peptide conjugate boosts antitumor efficacy through T cell engagement

Pan, Xue, and Yang et al. focused on improving efficacy of PD-1 blockade and PD-1 Ab-derived BiTEs (αPD-1). Using glycoengineering, a humanized anti-PD-1 was conjugated to a circular peptide (iRGD)2 that enhances deep tumor penetration, tumor cell internalization, and T cell bridging (via αv integrins, neuropilin, and PD-1). In syngeneic murine models, αPD-1-(iRGD)2 effectively penetrated and distributed in tumor tissue, expanded tumor-specific effector CD8+ T cells expressing stem- and memory-associated genes, and promoted TME remodeling. By mainly acting on PD-1+ CD8+ TILs, non-selective T cell activation and excessive TCR signaling were avoided.

Contributed by Katherine Turner

Neoantigen-augmented iPSC cancer vaccine combined with radiotherapy promotes antitumor immunity in poorly immunogenic cancers

An iPSC whole call cancer vaccine combined with radiotherapy (RT) improved CT26 tumor control compared to GM-CSF-adjuvanted iPSCs or fibroblasts by enhancing tumor cell apoptosis and cytotoxic T cell infiltration. The inclusion of RT engaged the STING pathway, as evidenced by i.t. mRNA. To expand the range of targeted antigens to include tumor-specific antigens, neoantigen-expressing iPSCs were engineered via AAV, which improved tumor control in the CT26 and 4T1 models, promoted infiltration of CD11c+ and granzyme B+ cells, and enhanced CT26 neoantigen-specific T cell responses, all of which were further enhanced by RT combination.

Contributed by Morgan Janes

Circadian control of tumor immunosuppression affects efficacy of immune checkpoint blockade

Fortin et al. showed that the circadian clock-mediated crosstalk between epithelial and immune cells in the intestine changes the tumor immune landscape, leading to immunosuppression. Genetic and environmental disruption of the epithelial cell clock resulted in the hyperactivation of pro-inflammatory cytokines and chemokines that promoted heightened inflammation, recruitment of neutrophils, and the subsequent development of MDSCs. PD-L1-expressing MDSCs rhythmically peaked in abundance during the early active phase, and the timing of anti-PD-L1 delivery enhanced treatment efficacy and outcomes in several tumor models.

Contributed by Shishir Pant

Principles of peptide selection by the transporter associated with antigen processing

Lee et al. performed cryo-EM experiments to analyze the structure of dimeric TAP while binding peptides. When TAP binds a peptide, the nucleotide-binding domains came closer together. The peptide N- and C-termini were stabilized mostly by backbone interactions in the peptide-binding pockets of TAP, although the pockets varied in size, depth, and electrostatic surface potential, allowing some side-chain specificity. To engage the binding pockets simultaneously, peptides had to be at least 8 residues long. End binding, while keeping the central positions in the transmembrane cavity, allowed for the binding of peptides of varying sequences and lengths.

Contributed by Maartje Wouters

Targeting BCL9/BCL9L enhances antigen presentation by promoting conventional type 1 dendritic cell (cDC1) activation and tumor infiltration

By analyzing published gene expression datasets, He, Wu, Liu, and Zhu et al. showed that BCL9 expression is negatively associated with tumor antigen presentation, immune cell influx, and overall survival of patients with colon and breast cancers and melanoma. Using mouse cancer models, scRNAseq analyses showed that Bcl9/Bcl9l deletion or pharmacologic inhibition of BCL9/β-catenin-driven Wnt transcription increased antitumor responses by boosting cDC1 activation and antigen presentation of tumor-derived antigens via TAK1/NFκB/IRF1-mediated upregulation of BTK, and CXCL9/CXCR3-mediated tumor influx and crosspriming of CD8+ T cells.

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.