Weekly Digests
‹ Back to March

CD4+ T cells help cDC1s provide CD8+ T cells with a license to kill

March 13, 2024

CD4+ T helper cells can stimulate CD8+ T cell responses by interacting with dendritic cells (DCs) through a process termed “DC licensing”. The conventional type 1 DC (cDC1) subset has been identified as the essential DC subtype for this process. Lei et al. investigated the mechanism of cDC1 licensing in human cells and the tumor microenvironment (TME) setting, and their data were recently published in Cellular and Molecular Immunology.

To assess the characteristics of “helped” and “non-helped” cDC1s, the researchers performed co-culturing assays in which human peripheral cDC1s were cocultured with naive (non-helped) or CD3/CD28-activated CD4+ T cells (helped). Gene set enrichment analysis revealed that in the helped cDC1s, cytokine/interleukin signaling, IFN-I response, and IFNγ signaling were upregulated. The researchers hypothesized that CD4+ T cells produced IFN-I in this setting. To test this, cDC1s were stimulated with activated CD4+ T cells or Poly(I:C). Expression of IFNα and IFNβ was upregulated in cDC1s stimulated by Poly(I:C), but not in those stimulated by activated CD4+ T cells. Additionally, IFNβ, but not IFNα, was upregulated in CD4+ T cells stimulated with both CD3 and CD28. Both results were consistent with CD4+ T cells being the source of IFN-I.

In myeloid cells, the STING pathway induces IFN-I production. STING signaling markers were measured in activated CD4+ T cells to determine whether their IFN-I production was also related to this pathway. Increased expression of STING pathway markers pSTING, pTBK1, and pIRF3 were detected at 24 hours after stimulation, and by 48 hours, IFNβ was produced. Using the STING agonist 2’3’-cGAMP in the assay showed increased protein levels of IFNβ, pSTING, and pTBK1, as well as IFN-I-stimulated gene products in the T cells. Furthermore, the agonist induced expression of markers of Th1 differentiation. STING inhibition, on the other hand, decreased expression of IFNβ in the T cells.

Lei et al. then determined whether cDC1s respond to IFN-I produced by CD4+ T cells by repeating the assay in the presence or absence of an anti-IFNα/β receptor 2 (IFNAR2)-blocking antibody. cDC1s upregulated expression of costimulatory molecules, chemokines, and antigen presentation pathway components after culture with IFN-I or after co-culturing with activated CD4+ T cells, which did not occur when IFNAR2 was blocked.

The researchers assessed whether IFNβ produced by CD4+ T cells impacted cDC1-mediated CTL responses in humans using an in vitro tumor antigen-specific CTL priming platform. CD8+ T cells were retrovirally transduced to express an HLA-A2-restricted MART-126-35 peptide-specific TCR. cDC1s were loaded with MART-115-40 long peptide or dead MART-1-expressing Mel526 cell debris to create antigen cross-presentation, and these cells were then exposed to activated MART-1 CD4+ T cells pretreated with or without IFNβ siRNA. Following long peptide loading of cDC1s, the proliferation of tetramer-positive CD8+ T cells did not differ between the siRNA-transfected and control CD4+ T cells, but granzyme B production was lower in the IFNβ1 siRNA-transfected setting. Loading of dead cells though resulted in a reduction of both proliferation and granzyme B when CD4+ T cells were treated with IFNβ siRNA, indicating that utilization of this natural antigen source was more sensitive to IFNβ stimulation from CD4+ T cells.

Activated CD4+ T cells upregulate the expression of CD40L, which can interact with CD40 on cDC1 for cDC1 licensing. To determine the contributions of IFNβ and CD40L in CD4+ T cell-mediated help, CD4+ T cells were edited by CRISPR-Cas9 to be deficient in IFNβ and/or CD40L, and were used in the activation coculture assay. Loss of IFNβ or CD40L in activated CD4+ T cells reduced expression of activation markers, MHC class II, and chemokines in cDC1s. However, only the IFNβ-deficient CD4+ T cells reduced the ability to upregulate key molecules involved in antigen cross-presentation, suggesting a role of IFN-I signaling in cDC1 licensing that is CD40-independent. In the CTL assay, loss of IFNβ or CD40L in activated CD4+ T cells reduced the priming of antigen-specific CD8+ T cells. CTL tumor cell killing capacity was also higher in cultures primed by helped cDC1s. In cultures primed with CD4+ T cells that were IFNβ- or CD40L-deficient, granzyme B production and CD107a expression decreased, and these CD8+ T cells were less capable of killing tumor cells.

Lei et al. then investigated the role of IFN-I-producing CD4+ T cells in cDC1 licensing in the human TME. In head and neck cancer tissue digests, IFNβ was detected in clusters of CD4+ T cells expressing Ki67, CXCL13, and PD-1. A cohort of ten scRNAseq datasets, including six types of human cancer, revealed that genes related to the IFN-I signature were highest expressed in the CD4+_ISG15, CD4+_CXCL13, and CD4+_MKI67 clusters. Gene signatures previously related to tumor reactivity in tumor-infiltrating lymphocytes (TIL) were enriched in the CD4+_CXCL13 and CD4+_MKI67 clusters, suggesting that the IFN-I-producing CD4+ T cells in the TME might be tumor-specific. A previous publication showed that a tumor-specific CXCL13+CD4+ T helper cell population in the TME interacts with LAMP3+ DCs, and the gene signature of this CXCL13+ T helper population was also enriched in the CD4+_CXCL13 and CD4+_MKI67 clusters. Another previous study detected various carcinoma ecotypes (CE), of which CE9 is correlated with good prognosis and is characterized by the combined presence of DCs, effector CD4+ and CD8+ T cells. The authors previously showed that the tumor-infiltrating DC signature in CE9 shares many characteristics with the “helped” cDC1 signature. Here, they detected that the gene expression signature of CD4+ T cells in CE9 is highly enriched in the detected CD4+_MKI67 population.

To determine the clinical relevance of these findings, the researchers performed survival analyses on TCGA data of seven cancer histologies. The CD4+_CXCL13 and CD4+_MKI67 signatures had a better prognostic value than other CD4+ T cell populations. Furthermore, to assess the predictive value of these populations for response to therapy, a melanoma cohort receiving anti-PD-1 therapy and an HPV+ oropharyngeal squamous cell carcinoma (OPSCC) cohort receiving standard-of-care therapies were analyzed. In melanoma, responders had higher CD4+_MKI67 and CD4+_CXCL13 signature levels than nonresponders. In OPSCC, when splitting patient groups based on immune response-positive (IR+) and -negative based on the presence of tumor-specific T cells among TILs, the IR+ patients had improved survival and higher levels of the CD4+_MKI67 and CD4_CXCL13 populations.

Together, these data suggest that IFN-I signaling induced by activated CD4+ T cells licenses cDC1 to induce antitumor CD8+ T cell responses, which might be associated with improved clinical outcomes. This helper role of CD4+ T cells in the TME may help overcome suboptimal priming of CD8+ T cells, and further research could shine more light on the role of various immunotherapies in improving this helper role and overcoming inhibition of this mechanism in the TME.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, lead author Yanling Xiao answered our questions.



What was the most surprising finding of this study for you?
In our pursuit to unravel the secrets behind cDC1's exceptional ability in tumor cell-associated antigen presentation, we explored the molecular processes. When activation of the IFN-I pathway occurred in the licensed cDC1, the source of IFN-I surprised us—it wasn't from presumed autocrine signaling, but originated from activated CD4+ T cells. The CD40/CD40L axis has been acknowledged as the primary player in cDC1 licensing. However, our discovery reveals that, despite intact CD40L/CD40 signaling, IFNβ-/- CD4+ T cells are unable to effectively license cDC1. Importantly, we find that MHC-I antigen cross-presentation in licensed cDC1 is mediated by IFN-I signaling rather than CD40. Through extensive exploration of the TME across multiple tumor types, we unveil a clinically favored subpopulation of IFN-I-producing CXCL13+CD4+ T-cells, reported to co-localize with helped/licensed cDC1.

What is the outlook?
Our findings advance cancer immunotherapy, showcasing cDC1 licensing in the human TME of favorable, T cell-infiltrated cancers. Transcriptomic evidence elucidates CD4+ T cell and cDC cross-talk, presenting a coherent scenario of CD4+ T cell help via cDC1 in the TME, mechanistically connecting with the historically known positive association of IFN-I signals. Our research consolidates evidence from recent impactful publications, providing a comprehensive understanding of CD4+ T cell and cDC1 interplay in the TME and paving the way for cancer immunotherapy advancements. Overall, this underscores the continual importance of research in this promising field. For that, we have recently been awarded funding from the Dutch Cancer Society (KWF) to bolster our efforts in identifying new targets for cancer immunotherapy in helped/licensed cDC1 within the TME.

What was the coolest thing you’ve learned (about) recently outside of work?
I am Chinese Dutch and lived in Japan for my Ph.D. Recently, I discovered raw fish roe at a delightful seafood stand in a charming town called Oegstgeest. Missing the Japanese dish called mentaiko, I decided to make it myself. Mentaiko is a delicacy consisting of roe sacs marinated in salt and chili pepper, enjoyed as an accompaniment to rice dishes. The process took seven days, and the final result brought me immense satisfaction, even though I am the sole enthusiast in the family. Delightful culinary creations have a unique ability to bring joy, not only to those who savor them, but also to the individuals who create these delectable dishes. The art of preparing food becomes a source of happiness, and when one has the skill to create dishes that bring profound happiness to many, it is a culinary achievement. The same holds true for a good piece of creative scientific work.

References:

Lei X, de Groot DC, Welters MJP, de Wit T, Schrama E, van Eenennaam H, Santegoets SJ, Oosenbrug T, van der Veen A, Vos JL, Zuur CL, de Miranda NFCC, Jacobs H, van der Burg SH, Borst J, Xiao Y. CD4+ T cells produce IFN-I to license cDC1s for induction of cytotoxic T-cell activity in human tumors. Cell Mol Immunol. 2024 Feb 21. 

In the Spotlight...

Entinostat, nivolumab and ipilimumab for women with advanced HER2-negative breast cancer: a phase Ib trial

In a phase 1b clinical trial, 12 women with advanced TNBC and 12 with HR-positive breast cancer were treated with entinostat (an HDAC inhibitor) + nivolumab + ipilimumab. Treatment was generally safe and well tolerated, with expected rates of AEs and no dose-limiting toxicities. In patients with TNBC, the ORR was 40% and the clinical benefit rate (CBR) was 60%, while in patients with HR-positive disease, the ORR was 25% and the CBR was 20%. PFS and OS rates were also better in patients with TNBC versus HR+ disease. Changes in myeloid cells, including evidence of increased antigen presentation, were linked to responses.

Contributed by Lauren Hitchings

Influence of microbiota-associated metabolic reprogramming on clinical outcome in patients with melanoma from the randomized adjuvant dendritic cell-based MIND-DC trial

The MIND-DC trial in patients with advanced melanoma showed that adjuvant natural DC (nDC) therapy induced Ag-specific immune responses, but did not provide a survival benefit. Digging deeper, Silva, et al. reported that despite randomization, there was a significant bias at baseline in serum metabolomics and fecal metagenomics that correlated with prognosis. At baseline, the nDC arm had significantly fewer commensals, such as Faecalibacterium prausnitzii, that highly correlated with 2-year RFS, as well as perturbations of primary biliary acids and lipids, suggesting host-microbial interactions influenced the nDC treatment response.

Contributed by Katherine Turner

The gene regulatory basis of bystander activation in CD8+ T cells

Watson et al. investigated “bystander activation”, in which T cells are activated by cytokines rather than TCR stimulation. Single-cell transcriptomics and chromatin accessibility profiling revealed that neonatal CD8+ T cells were more functionally diverse and more susceptible to IL-12- and IL-18-induced chromatin remodeling that opened up numerous AP-1 binding sites, enabling innate-like effector responses to non-specific pathogens. Most adult CD8+ T cells were less capable of such cytokine-induced chromatin remodeling, and instead expressed more Bach2, which blocks AP-1 binding. However, some innate-like CD8+ T cells could still be identified in both mice and patients into adulthood.

Contributed by Lauren Hitchings

An immunogenetic basis for lung cancer risk

Krishna et al. leveraged genetic and longitudinal clinical data from the FinnGen and UK Biobank, and found that heterozygosity at HLA-II loci is associated with reduced risk of lung cancer in both current and former, but not in never-smokers. Loss of heterozygosity (LOH) of the HLA-II loci favored the loss of alleles with larger neopeptide repertoires, and conferred substantial lifetime risk, independent of known clinical and genetic risk factors. Smoking affected HLA-II expressed in lung inflammatory macrophages and epithelial cells. Heterozygosity of amino acid sites within the HLA-II peptide binding groove was also associated with reduced risk of lung cancer.

Contributed by Shishir Pant

Targeting Dendritic Cell Dysfunction to Circumvent anti-PD1 Resistance in Head and Neck Cancer

Saito and Kono et al. demonstrated that Xcr1+ cDC1 dysfunction leads to reduced T cell priming in tumor-draining lymph nodes in an anti-PD-1-resistant syngeneic mouse oral carcinoma model (MOC). Moreover, in newly diagnosed patients with HNSCC treated with neoadjuvant anti-PD-1, CXCL9/10 expression and intratumoral cDC1 infiltration positively correlated with response. In the MOC model, an intratumoral cDC1 vaccine induced antigen-reactive T cell priming in the DLN, enhanced T cell infiltration, and overcame anti-PD1 resistance. Enforced CCL5 expression in the MOC model enhanced cDC1 tumor infiltration, restored antigen-specific responses, and sensitized tumors to anti-PD-1 treatment.

Contributed by Shishir Pant

STING Licensing of Type I Dendritic Cells Potentiates Antitumor Immunity

Wang et al. generated cGAMP-loaded STING-activating polymeric nanoparticles (Poly-STING) that induced a burst of, and then sustained STING signaling. In mouse models, intratumoral Poly-STING injection induced rejection of established and metastatic tumors, and memory to tumor rechallenge, which required cDC1 STING-IFN-I signaling and CD8+ T cells. Analyses of tumors from Poly-STING-treated WT and cDC1-deficient hosts led to the definition of a STING-cDC1 chemokine profile. This profile correlated with better survival in patients with melanoma and sarcoma. Neoadjuvant pembrolizumab (+/- chemotherapy) treatment of patients with NSCLC increased the STING-cDC1 profile.

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.