Weekly Digests
‹ Back to July

Resident memory T cells are ready to respond in neoadjuvant checkpoint blockade

July 13, 2022

Immune checkpoint blockade (ICB) has exciting potential in treating oral squamous cell cancers (OSCCs) in the neoadjuvant setting (i.e. prior to surgery). Although the identity of the responding T cells remains to be determined, tissue-resident memory (Trm) T cells, which play key roles in long-term immunity and can be rapidly engaged for effector responses, are an intriguing possibility. Recently reported in Cell, Luoma and Suo et al. identified Trms as the major responders to neoadjuvant ICB in an OSCC clinical trial.

In this phase 2 trial, 29 previously untreated OSCC patients were given anti-PD-1 therapy alone or with anti-CTLA-4 before surgical resection (within one week of the last dose of ICB). Therapeutic outcomes were promising – the majority of patients experienced reductions in tumor volume before surgery, and >80% of patients survived out to three years, relative to ~60% in historical cohorts. To investigate T cell responses, tumor biopsies and/or blood samples were collected pre-treatment, during treatment, and after surgery for single-cell RNA and TCR analyses.

To begin, Luoma and Suo et al. investigated T cell phenotypes within the OSCC tumors using scRNAseq. CD8+ T cells clustered into two main groupings: cells expressing GZMK and cells expressing GZMB. The GZMB cluster also expressed canonical Trm genes ITGAE and ZNF683 (encoding CD103 and HOBIT, respectively), along with cytotoxic and inhibitory gene sets, indicating this as a Trm population. scTCRseq showed that although all clusters had similar clonotype sizes before treatment, this ITGAE+ cluster was enriched for greater clonotype size after treatment, indicating expansion. Although cycling (MKI67) CD8+ and CD4+ T cells were also detected, their proportions increased only in the combination treatment (anti-PD-1 plus anti-CTLA-4) group.

The team next integrated bulk TCR sequencing with the scTCRseq data to locate the intratumoral TCR clonotypes that expanded following neoadjuvant ICB, which they called “treatment-expanded” (Tx-E) TCRs. Interestingly, only <1% of total clonotypes were identified as Tx-E, which the authors attribute to the short duration between treatment and resection/analysis. Most Tx-E TCRs (~59%) were found in pre-treatment tumor samples, but the remaining ~41% were only observed in tumors after ICB, suggesting de novo priming during neoadjuvant ICB. Notably, the Tx-E clonotypes were enriched in the ITGAE/GZMB T cell cluster, both in pre- and post-treatment samples. The proportion of Tx-E cells in this cluster was also higher with anti-PD-1 plus anti-CTLA-4 than with anti-PD-1 alone. In contrast, non-responding cells largely mapped to the GZMK cluster. Altogether, these results indicate that the ITGAE-expressing cells were readily invoked by ICB for treatment response, highlighting Trms as potential early responders to ICB.

Next, Luoma and Suo et al. directly compared gene expression between Tx-E and treatment-nonresponsive CD8+ T cells in tumors. Differentially expressed genes enriched in Tx-E cells encompassed effector, tissue resident, cytotoxic, and inhibitory pathways, and largely mapped to known Trm gene sets. A similar gene signature differentiated responding and non-responding CD4+ T cells; both CD8+ and CD4+ Tx-E T cells expressed effector and inhibitory genes including IFNG, LAG3, GZMA, and CXCL13. The researchers then developed a Tx-E gene signature, which they found correlated positively with treatment response and OS in an independent cohort of patients with urothelial cancer who were treated with anti-PD-L1.

To pinpoint the specific antigen targets of the Tx-E TCRs, the authors used TSCAN to screen 80 patient-derived CD8+ T cell TCRs against target cells presenting diverse self, mutant, and viral protein fragments on MHC-I. Through a GZMB reporter assay, six antigens were found targeted by TCRs that expanded after ICB treatment. These targets were a mix of self and tumor-associated antigens. Intriguingly, the team identified a novel epitope of the cancer-testis antigen MAGEA1 that induced a polyclonal response, which could have potential as an immunotherapy target.

Having analyzed intratumoral responses to neoadjuvant ICB, the researchers next considered whether these TCRs mapped to systemic T cell responses. In the blood, a CD8+ T cell cluster expressing CD38, HLA-DRA, and MKI67 increased in frequency over time. Interestingly, tumor Tx-E TCRs mapped largely to this population, and the previously identified tumor ITGAE/GZMB cluster also highly expressed CD38 and HLA-DR. Furthermore, TCR sequences from CD38+ HLA-DR+ CD8+ T cells sorted from blood matched tumor TCRs. Overall, the concordance between tumor and blood T cell phenotypes and TCR clonotypes responding to therapy suggests both a local and systemic response to ICB.

Because a significant portion (~41%) of Tx-E TCRs in the tumor was only detected post-therapy, the team investigated dynamics of these clones in the blood. The majority of these emergent TCRs were also found in blood after ICB, expanding ~2 weeks into treatment and declining by 10-12 weeks. These cells, and in fact all Tx-E clonotypes, expanded most significantly in the blood with the combination of anti-PD-1 and anti-CTLA-4 compared to anti-PD-1 alone – an interesting result given the known role of anti-CTLA-4 in supporting T cell priming.

Given the concordance between blood and tumor TCR clonotypes and phenotypes, the team finally investigated whether blood T cell features could potentially serve as biomarkers for ICB response. Strikingly, the CD38+ HLA-DR+ CD8+ T cell population in pre-treatment blood samples correlated with treatment response. These cells expanded during the course of treatment and displayed proliferative (Ki-67) and inhibitory (PD-1) markers, suggesting this activated T cell population as a potential biomarker for ICB efficacy. Pre- and on-treatment PD-1+ KLRG1- cells also strongly correlated with pathological response.

In conclusion, Luoma and Suo et al. identified Trm CD8+ T cells as early responders to neoadjuvant ICB in OSCC patients. Further investigating T cell phenotypes and responding populations could better tailor immunotherapy selection and delivery in future iterations. The identification of CD38/HLA-DR as potential T cell biomarkers can support this approach. Finally, the identification of a novel epitope capable of a CD8+ T cell response in OSCC provides another target for immunotherapy applications.

Write-up by Alex Najibi, image by Lauren Hitchings

Meet the researcher

This week, lead author Kay Wucherpfennig answered our questions. 

Lead author Kay Wucherpfennig.

What was the most surprising finding of this study for you? 
We investigated which immune cells respond early to pre-surgical (neoadjuvant) immune checkpoint blockade. The surprising finding was that those T cells that responded by clonal expansion expressed a tissue-resident memory program. We also found that there is an early systemic response to neoadjuvant immune checkpoint blockade characterized by a population of highly activated, proliferating T cells. Thus, neoadjuvant checkpoint blockade induces a local response in the tumor as well as a systemic response.  We think that the systemic response may be important in protecting against the outgrowth of metastases.

What is the outlook? 
We identified a novel biomarker in these studies. A substantial fraction of our patients had a pathological response (tumor necrosis evaluated by histology). We found that the activation state of circulating PD-1+ T cells was an important biomarker of pathological response. Importantly, the percentage of PD-1+ T cells was not associated with response, but the fraction of PD-1+ cells that were negative for the terminal differentiation marker KLRG1 strongly correlated with pathological response. We are now investigating the hypothesis that tissue-resident memory T cells are critical for cancer immunotherapy. This is relevant for many approaches to immunotherapy, including CAR T cells.

What was the coolest thing you’ve learned (about) recently outside of work? 
I am riding the Pan-Mass Challenge for Dana-Farber beginning of August. It’s 192 miles from Sturbridge to Provincetown at the tip of Cape Cod. 

References:

Luoma AM, Suo S, Wang Y, Gunasti L, Porter CBM, Nabilsi N, Tadros J, Ferretti AP, Liao S, Gurer C, Chen YH, Criscitiello S, Ricker CA, Dionne D, Rozenblatt-Rosen O, Uppaluri R, Haddad RI, Ashenberg O, Regev A, Van Allen EM, MacBeath G, Schoenfeld JD, Wucherpfennig KW. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell. 2022 Jul 1.

In the Spotlight...

Dissecting the treatment-naive ecosystem of human melanoma brain metastasis

Melanoma brain metastases (MBM) are common in patients with advanced melanoma, but the underlying biology is poorly understood. To address this gap, Biermann and Melms et al. compared multi-modal single cell transcriptomics of 22 treatment-naive MBMs and 10 extracranial melanoma metastases (ECMs; MBM and ECM were from different patients), coupled with functional validation in human and mouse models. MBM cells adopted a neuronal-like phenotype associated with increased chromosomal instability, and contained increased numbers of pro-tumorigenic macrophages and dysfunctional TOX+CD8+ T cells compared with ECMs.

Contributed by Katherine Turner

Tissue-resident memory CD8+ T cells possess unique transcriptional, epigenetic and functional adaptations to different tissue environments

Crowl and Hegg et al. used phenotypic, transcriptomic, and epigenomic analyses of CMV-specific (P14) T cells to demonstrate a role of tissue context in the generation and maintenance of critically important tissue-resident memory T cells (TRM). Although some TRM features were shared across tissues and were distinct from blood/spleen T cells, tissue-specific patterns emerged. Intraepithelial lymphocytes from the small intestine showed the greatest distinctions. TGFβ and chemokine axes can be drivers of specificity, and analysis of transcription factors revealed that Hic1 had opposing effects in different tissue contexts.

Contributed by Ed Fritsch

mTORC1 signaling in antigen-presenting cells of the skin restrains CD8+ T cell priming

Investigating the effects of mTORC1 signaling in vivo via deletion of the mTORC1 component raptor, Pelgrom and Patente et al. showed that mTORC1 regulated metabolism in an APC subset-specific manner, was required for effective cross-presentation by and maturation of cDC1s, and limited Langerhans cell activation. However, CD8+ T cell priming and IFNγ production in response to immunization were increased with loss of mTORC1, likely due to an accumulation of immunogenic EpCAM+ cDC1 subpopulation, which showed increased physical interactions with CD8+ T cells. These results point to mTORC as a potential target for immunotherapy.

Contributed by Lauren Hitchings

Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor-Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors

Kim et al. identified 39 T cell receptors (TCRs) specific for recurrent mutant p53 proteins in patients with solid tumors. In vitro and in vivo studies showed that these TCRs recognized tumor cells in a TP53 mutation- and HLA-specific manner and demonstrated antitumor activity. In 12 treated patients, ex vivo-expanded autologous TILs containing T cells reactive against TP53 neoantigens showed exhausted phenotypes and poor persistence. A patient with chemorefractory breast cancer treated with TCR-engineered PBL exhibited an improved immunophenotype, prolonged persistence, and an objective tumor regression (-55%) that lasted 6 months.

Contributed by Shishir Pant

WT1-specific TCRs directed against newly identified peptides install antitumor reactivity against acute myeloid leukemia and ovarian carcinoma

As a strategy to circumvent self-tolerance that limits WT1 recognition, van Amerongen et al. identified eight novel WT-1 peptides (distinct from WT-1 peptides currently targeted clinically) from the HLA-I ligandome of primary leukemia and ovarian cancer samples. High avidity CD8+ T cell clones were selected from healthy donors’ allogeneic T cell repertoire specific for the peptides presented by common HLA-I molecules. CD8+ T cells transduced with TCRs from those clones exhibited reactivity against WT1+ solid tumor cell lines and killed primary AML samples and ovarian carcinoma cell lines, but did not react to healthy cells or WT1- tumor cells in vitro.

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.