Weekly Digests
‹ Back to February

Tcf1+ progenitor-like T cell subsets sustain antitumor responses

February 13, 2019

Two articles, recently published back to back in Cell, identify important roles for T cells expressing Tcf1 (encoded by Tcf7), a transcription factor known to play roles in maintaining a relatively undifferentiated T cell state, promoting self-renewal and T cell memory. Kurtulus and Madi et al., working primarily with the MC38-OVA colon carcinoma model, focused their investigation on PD-1-CD8+ T cells, while Siddiqui et al., working with the B16F10 melanoma model, focused on PD-1+CD8+ T cells. Both teams defined a critical role for Tcf1+ T cells as a self-renewing precursor population critical to response to immunotherapy.

Kurtulus and Madi et al. began their investigation by analyzing bulk and single-cell RNA profiles of TILs to explore the dynamics of effector CD8+ T cell responses to checkpoint blockade. They found that following treatment with combination TIM-3 and PD-1 blockade in a murine OVA-MC38 colon carcinoma model, a variety of transcriptional changes occured, primarily within PD-1-TIM-3-CD8+ T cells (hereafter referred to as just PD1-CD8+ T cells). PD1-CD8+ T cells showed increased effector profiles and increased frequency following checkpoint blockade due to increased expansion in response to tumor-antigen stimulation. Adoptive transfer of this cell subset into MC38-OVA tumor-bearing RAG-/- mice showed that the transferred cells were able to maintain a population of PD-1-CD8+ T cells, while also producing a pool of PD-1+TIM3+CD8+ T cells. This suggested that PD-1-CD8+ TILs act as precursors with the potential to both self-renew and give rise to more differentiated cells.

Based on transcriptional programs, cell surface markers, and previously defined CD8+ T cell signatures, the researchers were able to identify three distinct subsets within PD-1-CD8+ T cells defined by expression of CD62L, Slamf7, and CXCR5: naive-like, memory-precursor-like, and effector-like. Effector-like and memory-precursor-like subsets both expanded in response to checkpoint blockade, contained tumor-antigen specific cells, and exhibited cytotoxic activity, though the memory-precursor-like cells exhibited higher polyfunctionality. Shifts in the proportions of subsets with checkpoint blockade and overlap in their gene expression profiles indicated that PD-1-CD8+ T cells likely follow a differentiation trajectory from naive-like, to memory-precursor-like, to effector-like. Hypothesizing that the memory-precursor-like PD-1-CD8+ T cell subset was largely responsible for maintaining long-lasting antitumor responses, the researchers identified high expression of the transcription factor Tcf1 in this subset. Targeted genetic knockout of Tcf7 in CD8+ T cells dramatically reduced the number and polyfunctionality of memory-precursor-like T cells and reduced the efficacy of checkpoint blockade. Finally, the gene signatures of the memory-precursor-like and effector-like subsets shared features with human TIL phenotypes that have previously been associated with better prognosis and response to checkpoint blockade in cancer patients, suggesting that these findings may be broadly applicable and relevant in the clinic.

In a separate study, Siddiqui et al., also identified an important role for Tcf1 (Tcf7), though their work largely focused on the PD-1+CD8+ T cell compartment. Siddiqui et al. first observed that adoptively transferred tumor-specific T cells lacking Tcf7 were less effective in controlling tumor growth in vivo than their Tcf7-expressing counterparts. The researchers found that most TILs from palpable untreated tumors were Tcf1+ and PD-1+. Therapeutic peptide vaccination expanded the Tcf1+PD-1+ TIL subset, concurrent with an expansion of Tcf1-PD-1+ TILs, which were more differentiated and frequently co-expressed granzyme B. When Tcf1+ cells were selectively ablated from vaccinated mice, Tcf1- T cells also decreased and tumors progressed, suggesting that the Tcf1+ subset sustains the Tcf1- subset in the context of therapeutic vaccination. Further, based on selective Tcf1 deletion and blockade of lymphocyte trafficking, the source of Tcf1- cells was found to be intratumoral Tcf1+ cells, suggesting that Tcf1+ cells are capable of expanding and differentiating on site, mediating the proliferative response to immunotherapy. Similar results were observed in the context of immune checkpoint blockade, suggesting that checkpoint blockade may rely not on the reversal of exhaustion in existing T cells, but on the expansion and differentiation of Tcf1+PD-1+ memory-like T cells within the tumor.

Like Kurtulus and Madi et al., Siddiqui et al. turned to clinical data to determine whether the subset they had identified was relevant in human patients. In peripheral blood from melan A peptide-vaccinated melanoma patients, the researchers were able to identify non-naive, tumor antigen-specific TCF1+PD-1+CD8+ T cells and TCF1-PD-1+CD8+ T cells with the same antigen specificity. Using publicly available single-cell RNAseq data for TILs from human melanoma, they confirmed that their subset of interest was present and had an expression profile similar to that observed in mice. In patients treated with checkpoint blockade, the fraction of TCF1+PD1+ cells was higher among CD8+ TILs than in patients who were not treated with checkpoint blockade. Using The Cancer Genome Atlas, Siddiqui et al. found that the combined expression of TCF7 and PDCD1 predicted improved patient survival.

Together, these studies by Kurtulus and Madi et al. and Siddiqui et al. suggest that immunotherapy acts on the less differentiated Tcf1+CD8+ T cells, which produce an expanded pool of more differentiated Tcf1-CD8+ T cells. While Kurtulus and Madi et al. interrogated PD-1- T cells and Siddiqui et al. focused on PD-1+ T cells, both identified the self-renewing and memory-stimulating effects of Tcf1 expression as critical to effective immunotherapy. Finally, given the potential of Tcf1+ memory or stem-like T cells to both self-renew and differentiate into Tcf1- effector-like cells upon treatment with checkpoint blockade, our understanding that checkpoint blockade reinvigorates existing exhausted cells may be replaced by a model in which checkpoint blockade depends on newly differentiated Tcf1- effector CD8+ T cells.

by Lauren Hitchings

References:

Kurtulus S., Madi A., Escobar G., Klapholz M., Nyman J., Christian E., Pawlak M., Dionne D., Xia J., Rozenblatt-Rosen O., Kuchroo V.K., Regev A., Anderson A.C. Checkpoint Blockade Immunotherapy Induces Dynamic Changes in PD-1(-)CD8(+) Tumor-Infiltrating T Cells. Immunity. 2019 Jan 15.

Siddiqui I., Schaeuble K., Chennupati V., Fuertes Marraco S.A., Calderon-Copete S., Pais Ferreira D., Carmona S.J., Scarpellino L., Gfeller D., Pradervand S., Luther S.A., Speiser D.E., Held W. Intratumoral Tcf1(+)PD-1(+)CD8(+) T Cells with Stem-like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy. Immunity. 2019 Jan 15.

In the Spotlight...

Clonal deletion of tumor-specific T cells by interferon-γ confers therapeutic resistance to combination immune checkpoint blockade

Pai et al. demonstrated in a TRAMP-C2 model that combination anti-CTLA-4 + anti-PD-1 controlled tumor growth in a high tumor burden (HTB), but not in a low tumor burden (LTB) state. Although T cells were less exhausted in LTB than in HTB, the combination therapy led to apoptosis of the dominant tumor antigen-specific T cells by activation-induced cell death via high IFNγ expression in LTB, which also limited effector memory formation. Patients with LTB advanced melanoma had lower response rates with combination therapy than with anti-PD-1 alone. These results suggest that response to combination therapy may vary by disease context.

Regulatory T cells mediate specific suppression by depleting peptide-MHC class II from dendritic cells

Akkaya and Oya et al. demonstrate with in vivo studies that Treg cells suppress the expansion of naive T cells in an antigen-specific manner. Mechanistically, Tregs bound peptide-pulsed dendritic cells (DCs) with more intensity than naive or activated T cells; following contact, Tregs removed the cognate peptide:MHCII complex from the surface of DCs via a process similar to trogocytosis, and thus prevented the activation of naive T cells recognizing the same antigen. Peptide:MHCII complexes with unrelated antigens were not removed from the surface of DCs, and therefore Tregs did not disable the capacity of DCs to activate other T cell responses.

CAR T cells targeting B7-H3, a Pan-Cancer Antigen, Demonstrate Potent Preclinical Activity Against Pediatric Solid Tumors and Brain Tumors

Majzner et al. showed that the immune checkpoint B7-H3 is highly and homogeneously expressed on many pediatric solid tumors (including sarcomas and brain tumors) and generated a CAR construct based on the humanized anti-B7-H3 antibody enoblituzumab. B7-H3 CAR T cells completely eradicated or significantly controlled established tumors in osteosarcoma, Ewing sarcoma, and medulloblastoma xenograft models, including metastases. In vitro and in vivo, CAR T cells killed tumor cells with high, but not low, expression of B7-H3, indicating a therapeutic window of tumor reactivity that might potentially limit toxicity.

IL2/anti-IL2 complex combined with CTLA-4, but not PD-1, blockade rescues antitumor NK cell function by regulatory T-cell modulation

In a lung tumor model, a complex of IL2/anti-IL2 (IL2Cx, which directs IL2 to NK and CD8+ T cells but not Tregs) and anti-PD-1 controlled tumor growth better than either monotherapy. In a B16F10-OVA melanoma model, IL2Cx delayed tumor growth, and this effect was boosted by either anti-CTLA-4 or anti-PD-1/PD-L1. CD8+ T cell infiltration, neoepitope-specific responses, and reinvigoration were increased by IL2Cx alone, and more so in combination treatments. The antitumor effect of both combinations was dependent on CD8+ T cells, and the effect of IL2Cx/anti-CTLA-4 was also dependent on NK cells, potentiated by Treg depletion.

WNT/β-catenin pathway activation correlates with immune exclusion across human cancers

Luke et al. analyzed the tumor microenvironment of solid tumors in The Cancer Genome Atlas and found that approximately one-third of them had poor T cell infiltration. Utilizing three approaches – analysis of somatic mutations or somatic copy number alterations, pathway target gene expression, and β-catenin protein levels – the researchers showed that WNT/β-catenin pathway activation inversely correlated with a T cell-inflamed tumor phenotype in 28 of 31 (90%) solid tumor types. This data suggest that WNT/β-catenin pathway activation is a possible mechanism of primary/intrinsic resistance to immunotherapy.

CD73 expression on effector T cells sustained by TGF-β facilitates tumor resistance to anti-4-1BB/CD137 therapy

Chen et al. demonstrated that the expression of CD73 (a cell-surface enzyme involved in adenosine-mediated immunosuppression) on host cells suppressed the antitumor effect of anti-4-1BB. Combination of anti-CD73 and anti-4-1BB led to tumor regression in mice with TGFβ-low tumors via a preferential expansion of intratumoral IFNγ-secreting CD73-CD8+ T cells and a decrease in Tregs. However, this treatment was ineffective in TGFβ-high tumors, as TGFβ maintained CD73 expression on T cells. Similar results were observed with anti-GITR or anti-OX40 treatments.

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.