Weekly Digests
‹ Back to March

Defining CD4+ T cells with a license to kill

March 10, 2021

The helper function of CD4+ T cells in antitumor immune responses has been characterized, but much less is known about the role of cytotoxic CD4+ Th cells (Th-CTX) in tumors. Cachot, Bilous, and Liu et al. investigated the presence, phenotype, and cytolytic function of tumor-specific Th-CTX using single-cell transcriptomics, peptide–MHC-II multimers, and a newly developed high-throughput single-cell cytotoxicity assay. Their exciting results were recently published in Science Advances.

The researchers started by mining publicly available single-cell RNAseq datasets of tumor-infiltrating T cells in advanced melanoma for CD4+ T cell subsets. Unsupervised clustering of more than 2,000 CD3+CD4+ T cells revealed four main clusters, which were annotated as regulatory, memory, cycling, and cytotoxic clusters based on gene expression. The cytolytic cluster was enriched for lytic granule genes and genes previously associated with effector and cytolytic functions in CD4+ T cells, such as CRTAM, NKG7, KLRG1, and CST7. This cluster was also enriched for genes involved in the cellular response to IFNγ and chemotaxis, with upregulation of the chemokines CCL5, CCL4, and CCL4L1/2. No clear single transcription factor appeared to be critical in the cytotoxic cell population, suggesting that multiple factors may be involved in defining this phenotype.

To further investigate the cytolytic features of the Th-CTX population, the researchers used peptide–MHC-II multimers to isolate NY-ESO-187-99/DR7-specific CD4+ T cells from peripheral blood, and from tumors and tumor-infiltrated lymph nodes from patients with melanoma. These multimer-positive T cells were found in 8/9 blood samples and 5/8 tumor samples. The peripheral cells showed higher granzyme B expression than those in the tumor, while tumor-associated cells expressed higher perforin, granzyme A, and BLIMP-1 levels.

Direct cytolytic antitumor activity of Th-CTX would require tumoral MHC-II expression, which was investigated by assessing HLA-DR by fluorescent IHC staining of melanoma tissue sections. Five out of 18 cases had HLA-DR+ tumor cells, and in vitro experiments confirmed that both IFNγ and TNFα could elevate MHC-II expression on melanoma cell lines. These findings demonstrate that interaction between Th-CTX and tumor cells in the tumor microenvironment is possible.

To assess the cytolytic capacity of Th-CTX, the researchers isolated and expanded multimer-positive sorted cells from patient samples and performed a 51chromium release assay. Each clone was tested in the presence of peptide-loaded tumor cells that were either pretreated with IFNγ or stably transduced with CIITA to increase MHC-II expression. Up to 46% lysis occurred in CIITA-transduced tumor cells, but less lysis was observed in IFNγ-treated and wild-type cells, reflecting differences in MHC-II expression. Similar results were found with Mage-A3 epitope-specific T cells.

The researchers then developed a high-throughput single-cell tumor recognition assay to monitor interactions between effector and target cells and evaluate cytotoxicity at the single-cell level. The data were analyzed using deep learning tools, with an algorithm that enabled the quantitation of various effector to target cell ratios and the target cell lysis specificity. Although cytolysis started after a few minutes of coculture, it persisted 15-20 hours, with an average killing time of 5.3 hours; this is slower than the average killing time of 2.4 hours by CD8+ cells in similar assays. There was evidence of serial killing activities and a trend toward a higher lytic capacity when there was an excess of targets, which is in line with previous work on the cytolytic capacities of CD8+ T and NK cells.

To look into the pathways that induced the cytolytic activity of the Th-CTX, the researchers assessed cytokines and other molecules produced by antigen-specific Th-CTX and Th CD4+ T cell clones after coculture with peptide-loaded target cells. Th-CTX and Th cells produced little perforin, granzyme K, and granzyme M, but high concentrations of granzyme A. Additionally, the Th-CTX cells produced significantly more granzyme B and granulysin than Th cells. The importance of lytic granules for these cells' killing capacity was confirmed in a coculture experiment performed in the presence of concanamycin (a compound that degrades perforin present in vacuoles), which resulted in a decrease of lysis. The lysis was found to be contact- and granzyme B-dependent.

Next, the TCR α and β chains of 32 NY-ESO-187-99/DR7-specific CD4+ T cell clones isolated from blood were assessed. The clones showed diverse, clonally distributed TCRs in both Th and Th-CTX, with high variability between patients. Both shared and distinct TCR α and β chains were identified in the two cell populations, with similar membrane expression of TCRαβ in all clones, suggesting the TCR expression had minimal impact on the functional differences between these populations.

To determine specific surface markers of the Th-CTX, the researchers used the scRNAseq data. SLAMF7 was identified as predominantly expressed by cells in the cytolytic cluster, while PD-1 was expressed mainly in the cycling and cytolytic clusters. Assessing the protein levels on blood and tumor-associated NY-ESO-187-99/DR7-specific CD4+ T cells, both SLAMF7 and PD-1 were detected directly ex vivo, with lower expression of PD-1 on peripheral cells and more double-positive cells in the tumor. The expression of SLAMF7 could be induced in vitro by IL-12 or the histone deacetylase inhibitor Entinostat. Although SLAMF7 expression was favorably associated with a good clinical outcome in the TCGA dataset, further studies are needed to link this effect to Th-CTX.

To establish the roles of SLAMF7 and PD-1 in the cytolytic function of the Th-CTX, the 51chromium release assay was repeated with the addition of either an anti-PD-1 or an agonistic anti-SLAMF7 antibody. The percentage of specific lysis was not enhanced by PD-1 signaling blockade, but SLAMF7 stimulation slightly increased lysis. Finally, Th-CTX were found in scRNA-seq datasets from breast, head and neck, and hepatocellular cancers. However, SLAMF7 was only upregulated in melanoma, breast cancer, and hepatocellular carcinoma – not in head and neck cancer.

In summary, the data presented here demonstrate the presence of an interesting tumor-specific CD4+ T cell subpopulation with cytolytic capacity in multiple tumor types and characterize their phenotypic and functional properties, supporting the potential of MHC-I-independent targeting. Further, these studies reveal SLAMF7 agonism on Th-CTX cells as a potential new immunotherapy target.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, lead author Camilla Jandus answered our questions.

What prompted you to tackle this research question?

Immunotherapy, an innovative treatment strategy aimed to boost the immune response against cancer, has significantly changed management of patients with cancer. However, many patients still do not respond to treatment or develop resistance. To date, most studies have focused on boosting and re-invigorating cytotoxic CD8+ T cell functions, while still little is known about the role of CD4+ T cells in antitumor immunity, and particularly in their direct contribution in eliminating tumors. We were really interested in combining innovative approaches down to the single cell level to investigate whether human CD4+ T cells can directly eliminate tumor cells. Further we aimed at identifying the determinants that are linked with their antitumor functions, to be exploited in cancer patients.

What was the most surprising finding of this study for you?

CD4+ T cell cytotoxicity has been reported in the context of viral infections, vaccination, and tumor mouse models, but still little is known regarding the presence and overall frequency of these cells in human tumors. I was surprised to observe very frequent and efficient CD4+ T cell killing of targets in the picowells of the real-time nanobiosensor that we developed together with Professor Altug at the EPFL. Even more surprising was the fact that when a single CD4+ T cell was outnumbered by target cells, serial engagement in multiple killing activities was observed.

What was the coolest thing you’ve learned (about) recently outside of work?

In the recent cold winter days in the Swiss mountains, I discovered the beauty of the Mpemba effect, a phenomenon that refers to the fact that, contrary to natural prediction, in certain circumstances, hot water freezes faster than cold water. Although the theoretical basis of this physical paradox, initially noticed by Aristotle, remains a matter of controversy, the practical result of it is that during icy days such as the ones of last February, you can easily create ice rainbows using a bottle of boiling water. Seeing is believing!

References:

Cachot A., Bilous M., Liu Y.C., Li X., Saillard M., Cenerenti M., Rockinger G.A., Wyss T., Guillaume P., Schmidt J., Genolet R., Ercolano G., Protti M.P., Reith W., Ioannidou K., de Leval L., Trapani J.A., Coukos G., Harari A., Speiser D.E., Mathis A., Gfeller D., Altug H., Romero P., Jandus C. Tumor-specific cytolytic CD4 T cells mediate immunity against human cancer. Sci Adv. 2021 Feb 26.

In the Spotlight...

Targeting telomerase with an HLA class II-restricted TCR for cancer immunotherapy

Dillard et al. reported on the isolation and preclinical efficacy of novel hTERT-specific CD4+ T cell-derived TCR (Radium-4) from a clinically responding pancreatic cancer patient vaccinated with a long hTERT peptide. Radium-4 TCR-expressing CD4+ and CD8+ T cells recognized hTERT in the context of HLA-DP04 and HLA-DP03, produced inflammatory cytokines and demonstrated cytotoxicity against melanoma cell lines engineered to present TERT peptides grown in 2D and 3D cultures. Radium-4 TCR-transduced T cells reduced tumor growth and prolonged survival in melanoma xenograft models and lacked cytotoxicity against hematopoietic stem cells.

Contributed by Shishir Pant

Heterologous arenavirus vector prime-boost overrules self-tolerance for efficient tumor-specific CD8 T cell attack

Due to globally low seroprevalence in humans and elicitation of CTL responses of exceptional magnitude and longevity, attenuated arenavirus vectors that express tumor antigens are attractive cancer vaccine candidates. Bonilla et al. primed tumor-bearing mice with tumor antigen-expressing attenuated arenavirus vectors, and then switched to distantly related arenavirus vectors for the boost. This switch resulted in post-boost avoidance of secondary T cell responses to immunodominant vector epitopes, enhanced frequency of tumor-recognizing CTLs, increased survival with immunological memory, and the breaking of self-tolerance to tumor antigens.

Contributed by Margot O’Toole

Generating tumor-selective conditionally active biologic anti-CTLA4 antibodies via protein-associated chemical switches

Chang et al. developed novel, conditionally active biologic (CAB) anti-CTLA-4 mAbs by selecting variants with reversible, pH-dependent binding to CTLA-4 at pH 6.0 (acidic TME) but not at pH 7.4 (healthy tissue). CAB selectivity was characterized as responsive to physiological levels of bicarbonate and hydrogen sulfide as non-covalent protein-associated chemical switches (PaCS). CAB antitumor efficacy was similar to an ipilimumab analog (IpA), but (in combination with anti-PD-1) resulted in dramatically less GI toxicity and peripheral T cell activation compared to IpA in an NHP toxicology study. PaCS can be applied to many Ab formats to improve the therapeutic index.

Contributed by Katherine Turner

CRISPR/Cas9-engineered universal CD19/CD22 dual-targeted CAR-T cell therapy for relapsed/refractory B-cell acute lymphoblastic leukemia

In an open-label phase I study, Hu et al. evaluated the safety, feasibility, and efficacy of off-the-shelf CD19/CD22 dual-targeting allogeneic CAR T cells (CTA101) with a CRISPR/Cas9-disrupted TRAC region and CD52 gene. All six enrolled relapsed/refractory B-cell ALL patients received one infusion of CTA101 within 8 days of enrollment, showed cytokine release syndrome, and lacked CRISPR gene editing-associated genotoxicity, chromosomal translocation, neurotoxicity, or graft-versus-host disease. On Day 28, five out of six patients achieved complete remission, and all patients who had a response achieved MRD-negative remission.

Contributed by Shishir Pant

FcγRIIB is a T cell checkpoint in antitumor immunity

Farley et al. detected FcγRIIB on activated, exhausted CD8+ T cells in tumors, spleens, and draining lymph nodes (dLN) of mice inoculated with a melanoma cell line. In Fcgr2b-/- compared to wild-type mice, tumor volume was lower, CD8+ TEM cells increased in tumors and spleens, and PMA+ionomycin-induced IFNγ+TNF+ cells increased in the tumor, spleen, and dLN. Adoptively transferred antigen-specific Fcgr2b-/- CD8+ T cells preferentially expanded in tumors, and antigen restimulation ex vivo expanded Fcgr2b-/- CD8+ antigen-specific IFNγ+ TNF+ T cells in dLNs and spleens. FcγRIIB+CD8+ T cells were detected in patients with metastatic melanoma.

Contributed by Paula Hochman

A Potent Cancer Vaccine Adjuvant System for Particleization of Short, Synthetic CD8+ T Cell Epitopes

He et al. developed a peptide-binding vaccine carrier based on a cobalt porphyrin-phospholipid liposome containing QS-21 (a saponin adjuvant) and PHAD (a TLR4 agonist) (altogether, CPQ). CPQ readily bound the gp70 peptide A5 and improved in vitro APC uptake into MHC-I-containing endosomes, and in vivo DC accumulation in the dLN. Prophylactic A5-CPQ immunization induced robust A5-specific T cell responses, inhibited gp70-expressing tumors, and was nontoxic in CD-1 mice. In a therapeutic setting, A5/CPQ regressed CT26 tumors and eliminated lung metastases. Immunization with CPQ was used to functionally screen candidate neoantigen peptides.

Contributed by Alex Najibi

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.