Weekly Digests
‹ Back to September

Tumor-specific and tissue-resident memory T cells may hold the key in TNBC

September 7, 2022

Investigating the importance CD39+CD8+ T cells in prognosis and in the success of immune checkpoint blockades for breast cancer, Lee et al. examined the heterogeneity of CD8+ T cells obtained from different compartments, including primary tumors, metastatic lymph nodes (mLNs), and peripheral blood, and characterized cellular phenotypes and functionalities of T cells using data from multicenter cohorts of patients with breast cancer who underwent curative aimed-surgery. Their results were published in Science Immunology.

To begin, the team looked at the composition of immune cells in peripheral blood and tumor tissues from 131 patients with early breast cancer. Among CD45+ cells, T cells were the most abundant cell type in tumors. Among T cells, the percentage of CD8+ cells was higher in tumors than in blood, as was the percentage of CD8+ T cells expressing CD69 and CD103 – markers of tissue-resident memory T cells (TRM). CD103 was later found to be exclusive to CD69+CD8+ T cells, and CD103 was subsequently used to define TRM.

Investigating the antigen specificity of T cells within tumors, the researchers first identified virus-specific bystander T cells. While bystander T cells recognizing influenza A virus (IAV)- were enriched in tumors over peripheral blood, those recognizing human cytomegalovirus (HCMV) were not, possibly because a higher percentage of IAV-specific T cells were CD69+CD103+ TRM. Next, the team identified T cells recognizing NY-ESO-1, which is frequently expressed in triple-negative breast cancer (TNBC). These cells could be found in the tumors and blood of patients with TNBC. Within the tumor, but not the peripheral blood, NY-ESO-1-specific CD8+ T cells were enriched for TRM relative to all CD8+ TILs.

Next, the researchers evaluated expression of CD39, which is typically expressed in tumor antigen-specific T cells. Here, CD39 expression was increased among TRM vs non-TRM. Sorting CD8+ TILs into subpopulations based on tissue residency and tumor antigen specificity, the researchers defined three subpopulations: CD39+ TRM, CD39- TRM, and non-TRM. Gene clustering analysis, Gene Ontology (GO) biological pathway analysis, and gene set enrichment analysis together revealed features of functional exhaustion in CD39+ TRM, including upregulated expression of genes related to exhaustion, immune checkpoints, effector functions, and activation, and downregulation of genes associated with naive or central memory CD8+ T cell phenotypes. Genes upregulated in CD39+ TRM were also more characteristic of previously identified TRM signatures, whereas CD39- TRM were more effector memory T cell-like. Upon activation, CD39+ TRM produced less IFNγ, TNFα , and IL-2, and were less polyfunctional compared to other subpopulations. Similar results were observed in cells from mLNs.

Turning their attention towards clonality, Lee et al. performed TCR analysis and compared CD8+ T cell populations between primary tumors, mLNs, and peripheral blood. Defining cells as naive, (TN), central memory (TCM), effector memory (TEM), or terminally differentiated effector memory (TEMRA), the team found that TEM were predominant in tumors and mLNs. Further, many TRM were detected within the TEM population. Within each compartment, some clonotypes overlapped between CD39+ TRM, CD39- TRM. and non-TRM, with minimal effects of contaminating minor clonotypes on TCR clonotype analysis. Between compartments, anywhere from 9-62% of clonotypes found in tumors or mLNs were also detected in other compartments (intercompartmental), with over 80% of intercompartmentally overlapping clonotypes found in peripheral blood. Clones that overlapped between tumors and mLNs were mainly identified within the same subpopulations (CD39+ TRM, CD39- TRM, or non-TRM), and mainly overlapped with TEM as highly expanded (large) clones in the blood. Further, blood TEM clonotypes were distributed among various CD8+ T cell subpopulations, including CD39+ and CD39- TRM, suggesting that blood TEM may differentiate into TRM in tumors and mLNs.

Using a coculture system, Lee et al. confirmed that CD39+ T cells recognized tumor antigens and were reactive against tumor cells. Tumor-reactive (upregulated 4-1BB) TRM were then sorted and TCR sequenced. Mapping these cells back to the patient, the researchers found that tumor-reactive 4-1BB+CD39+ TRM clonotypes with inter-subpopulation clonal overlap in culture were readily detected in blood TEM cells as large clones and broadly distributed among various subpopulations in tumors and mLNs compared with those without inter-subpopulation clonal overlap, confirming systemic clonal overlap.

Lee et al. then examined CD39+ TRM enrichment in early breast cancers with different molecular subtypes: HR+/HER2-, HER2+, and TNBC. While the portion of CD8+ among CD3+ cells and CD103+ among CD8+ cells was consistent across molecular subtypes, the portion of CD39+ among CD8+ cells varied, with the highest enrichment of CD39+ cells in TNBC and the lowest in HR+/HER2- breast cancer. Applying a gene signature for CD39+ TRM to data from The Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) and The Cancer Genome Atlas (TCGA) also showed that that CD39+ TRM were highest in TNBC and lowest in HR+/HER2- breast cancer, including in a metastatic tissue cohort. They also found that while the CD39+ TRM score did not correlate with survival in HR+/HER2- or HER2+ breast cancer cohorts, it did strongly correlate with better cancer-specific survival in TNBC, indicating that CD39+ TRM may directly contribute to antitumor immune responses.

To determine whether CD39+ TRM could be reinvigorated by checkpoint blockade, the researchers performed ex vivo functional analysis assays using CD8+ TILs from a patient with TNBC and NY-ESO-1-specific CD8+ T cells. Here, stimulation with NY-ESO-1 peptides induced proliferation and cytokine release in CD39+ TRM, but not CD39 TRM or non-TRM, and these effects were enhanced with anti-PD-1 and with the addition of anti-CTLA-4. Similar results were observed when CD8+ TILs from another patient with TNBC were cultured with autologous EPCAM+ tumor cells. While restoration of functionality with checkpoint blockade was limited in samples from HR+/HER2- breast cancer, a range of results were observed in samples from patients with HER2+ breast cancer or TNBC, and restoration correlated with enrichment of CD39+ TRM, suggesting that these cells are likely a responding subpopulation that can be functionally restored.

Finally, comparing single-cell RNAseq data from a clinical trial of neoadjuvant anti-PD-1 in patients with breast cancer to their bulk RNAseq data, the researchers found that upregulated genes in expanding CD8+ TILs were also enriched in CD39+ TRM over CD39- TRM. Similarly, applying their CD39+ TRM signature against public scRNAseq dataset from a trial of anti-PD-L1 in patients with advanced TNBC, they found upregulation of CD39+ TRM signature genes in CXCL13+CD8+ T cells, which were associated with response to PD-L1 blockade.

Together, these results show that enrichment of CD39+ TRM, especially those that clonally overlap across compartments, may contribute to survival and response to checkpoint blockade in some patients with breast cancer. These findings may be useful in evaluating therapeutic options for such patients.

Write-up and image by Lauren Hitchings

References:

Lee YJ, Kim JY, Jeon SH, Nam H, Jung JH, Jeon M, Kim ES, Bae SJ, Ahn J, Yoo TK, Sun WY, Ahn SG, Jeong J, Park SH, Park WC, Kim SI, Shin EC. CD39+ tissue-resident memory CD8+ T cells with a clonal overlap across compartments mediate antitumor immunity in breast cancer. Sci Immunol. 2022 Aug 26.

In the Spotlight...

Differences in CD80 and CD86 transendocytosis reveal CD86 as a key target for CTLA-4 immune regulation

Kennedy, Waters, and Rowshanravan et al. showed that CTLA-4 captured its tagged ligands from opposing cells to mediate transendocytosis (TE) into CTLA-4+ cells. CD86 dissociated from CTLA-4 in a pH-dependent manner to enable CTLA-4 recycling, whereas CD80 stayed bound to CTLA-4, which was ubiquitylated and trafficked via late endosomes/lysosomes to prevent CTLA-4 recycling. CTLA-4 recycling defects due to CD86 TE disruption caused by deficiency of a key protein trafficking regulator, LRBA protein, or by specific CTLA-4 mutations led to T cell dysregulation in vitro, and associated with clinical autoimmunity.

Contributed by Paula Hochman

Fc-γ receptor-mediated crosslinking co-defines the immunostimulatory activity of anti-human CD96 antibodies

Rogel et al. engineered the Fc domain of anti-human CD96 IgGs that inhibited CD96 binding to CD155 (also a TIGIT and CD226 ligand). Anti-CD3-induced T cell proliferation was co-stimulated by immobilized, FcγR-crosslinked, and soluble anti-CD96 IgG1s trans-crosslinked by FcγRI+ accessory cells. Functional, ELISA, and RNAseq analyses showed that anti-CD96 IgG1 directly costimulated T cell proliferation and effector function, and overcame Treg suppression. Anti-CD96 IgG1 costimulated TIL proliferation, and TGCA data showed that CD96 expression correlated with survival in HPV+ head and neck squamous cell carcinoma.

Contributed by Paula Hochman

Multifunctional nanoparticle potentiates the in situ vaccination effect of radiation therapy and enhances response to immune checkpoint blockade

To improve the antitumor immunity of in situ vaccination using radiation therapy (RT) combined with immune checkpoint blockade (ICB), Zhang, Srirameni et al. tested a scalable nanoparticle consisting of polylysine, iron oxide, and CpG (PIC) that was designed to down-modulate TME suppression. When combined with RT and ICB, PIC enhanced in situ vaccine RT efficacy, and primed a potent systemic antitumor response with cures and long-term memory in several syngeneic “cold” tumor models. PIC addition increased tumor antigen presentation by DCs, increased the M1:M2 TAM ratio, antagonized the RT-induced increase in Tregs, and stimulated a type 1 interferon response.

Contributed by Katherine Turner

Outcomes with adjuvant anti-PD-1 therapy in patients with sentinel lymph node-positive melanoma without completion lymph node dissection

To assess the real-world benefits of immediate (IM) complete lymph node dissection (CLND) in patients SLN+ melanoma treated with systemic adjuvant therapy, Eroglu, Broman, and Thompson et al. retrospectively analyzed 462 patients. In the subset (n=386) with adjuvant anti-PD-1, the 24-month RFS was comparable among patients with (n=60) and without (n=326) IM CLND and to prior adjuvant anti-PD-1 trials with high rates of IM CLND. Patients without IM CLND had a significantly higher rate of locoregional relapses, and those with SLN tumor deposit >1 mm, stage IIIC/D, and ulcerated primary had worse RFS. Secondary adjuvant therapy did not affect second relapse in patients without IM CLND.

Contributed by Shishir Pant

Image-guided intratumoral immunotherapy: Developing a clinically practical technology

Som et al. reviewed the physics, chemistry, and mechanics of using imaging for intratumoral (i.t.) delivery of immunotherapy/immunoadjuvants. I.t. delivery may aid in converting immune-cold tumors to a hot phenotype to improve efficacy. Ultrasound and CT imaging are the most used methods for precision drug delivery and monitoring drug distribution; delivery tracing reagents are critical. Using microparticles, hydrogels, and RNA vector carriers for drug delivery may improve local drug distribution and offer controlled sustained release. This can create higher local drug concentrations, which may improve efficacy, and, importantly, reduces the need for repeated treatment.

Contributed by Maartje Wouters

Natural Antibodies Alert the Adaptive Immune System of the Presence of Transformed Cells in Early Tumorigenesis

Rawat et al. demonstrated that natural Abs (Nabs) are critical for the early-stage recognition of and immune cell activation against adoptively transferred immunogenic and nonimmunogenic neoantigen-expressing cells and transplanted and spontaneous tumors. Adoptively transferred neoantigen-expressing cells persisted in NAb-deficient mice, whereas they were eliminated in wild-type (WT) mice. Restoration of the NAb repertoire in NAb-deficient mice with either WT B cells or naive serum elicited the recognition and elimination of neoantigen-expressing cells in a T- and B cell-dependent manner. The antitumor immune response was dependent on NAbs and not on the Ag-presenting property of B cells.

Contributed by Shishir Pant

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.