Weekly Digests
‹ Back to March

CD5 on DCs brings antitumor T cell responses to life

March 1, 2023

To induce effective antitumor T cell responses in response to immune checkpoint blockade (ICB) treatment, effective priming by dendritic cells (DCs) is essential. However, the characteristics of effective DC responses during ICB remain largely unclear. Having previously shown that migratory CD5+ DCs in the skin can prime CD4+ and CD8+ T cells more effectively than other DC subsets, He et al. assessed the role of this DC subset in antitumor immunity and ICB therapy responses. Their results were recently published in Science.

The researchers sorted myeloid cells from tumor-draining lymph nodes (TDLNs) and unaffected lymph nodes of patients with metastatic melanoma, and assessed these cells by single-cell RNAseq. CD5 expression was mostly detected on cDC2s in the unaffected LNs. Confirming these data, flow cytometry and CyTOF analysis confirmed the presence of CD1c+CD5+ DCs in uninvolved LNs. Assessing the prognostic role of CD5 using TCGA data showed that CD5 expression and the CD5+ DC signature were associated with improved prognosis in melanoma, lung squamous cell carcinoma, sarcoma, breast cancer, cervical squamous cell carcinoma, and endocervical adenocarcinoma.

In vitro experiments showed that CD5+ DCs had enhanced capacity to activate and induce proliferation of naive CD4+ and CD8+ T cells, resulting in higher production of IFNγ and TNFα by the T cells. To determine whether CD1c+CD5+ DCs were capable of reactivating CD8+ T cells specific to influenza matrix protein M1 (Flu-M1), DCs were isolated from an HLA-A2+ donor, and loaded with a peptide containing the HLA-A2-restricted epitope from Flu-M1. Coculturing these DCs with autologous peripheral CD8+ T cells resulted in higher antigen-specific T cell frequencies than coculture with other DC populations. Similarly, CD5+ DCs more efficiently activated MHC-II-restricted viral CD4+ T cell responses.

To determine the impact of CD5 expression levels, dermal CD1c+ DCs were sorted based on the number of CD5 molecules. Coculture experiments showed that the most effective priming of CD8+ T cells was induced by CD5hi DCs, resulting in a higher frequency of CD8+ T cells producing IFNγ, granzyme B, and perforin. CD5hi DCs also primed a higher frequency of multifunctional T cells expressing two or three effector molecules than CD5int or CD5- DCs. CD5hi DCs also induced higher proliferation of CD4+ T cells and IFNγ production than the other DC subsets. These effects were inhibited when a blocking CD5 antibody was added to the culture. On the other hand, when CD5 was upregulated on CD5- DCs by CRISPR gene activation, naive T cell proliferation and priming increased, suggesting that CD5 expression on DCs plays an essential role in these processes.

In contrast to the human data, CD5 was found on both cDC1 and cDC2 subsets in mice. The researchers generated conditional CD5 knockout mice using CRISPR-Cas9 to delete CD5 specifically in DCs (CD5ΔDC). To determine whether CD5 expression on DCs was required to induce antitumor immune responses in vivo, the MCA-induced syngeneic sarcoma cell line engineered to express ovalbumin (OVA) (MCA1956-mOVA) was used. While 14/15 WT mice eliminated tumors, only 2/11 CD5ΔDC mice rejected the tumor, and tumor-reactive CD8+ T cells expanded only in control mice.

To assess the implications on CD4+ T cell responses, antigen-specific T cell activation assays were performed using co-culturing of CD5-deficient DCs or control cDC2s with purified T cells from OT-II mice in the presence of a long peptide containing the MHC-II-restricted epitope. This showed that the CD5- DCs were less efficient at activating and inducing the proliferation of OT-II T cells.

Next, the researchers assessed the effects of CD5 expression on DCs for the efficacy of ICB therapy. Parental MCA1956 tumors grew progressively in control and CD5ΔDC mice, but only control mice responded to anti-PD-1 treatment. Similar results were obtained for anti-CTLA-4 therapy. Using the MC38 colorectal cancer model, the researchers found that the frequency of CD8+ T cells reactive to mutated epitopes from the mutated ADP-specific glucokinase (a neoantigen in MC38) was lower in tumors of CD5ΔDC mice, suggesting reduced priming.

In mice with CD5-deficient T cells (CD5ΔT), the growth of MCA1956 tumors was comparable with control mice. However, most of the control mice (13/16) responded to anti-PD-1 treatment, while none of the 14 CD5ΔT mice rejected the tumor. Similarly, in mice with CD5-deficient T cells, MCA 1956-mOVA tumors were not spontaneously rejected, and these mice had reduced numbers of OVA-specific CD8+ T cells.

ICB treatment in control mice did not alter the frequencies of DCs and macrophages, but it did upregulate the frequency of CD5+ DCs in the tumor and TDLN. CD5+ DCs were mainly found in the LN after treatment, and had lower levels of CCR7, suggesting the increase was not due to increased migration of these DCs. However, in vitro experiments showed that CD5+ DCs were more resistant to apoptosis than CD5- DCs in the presence of anti-PD-1.

In human T cells isolated from LNs, a larger fraction of T cells was found to express CD5 in LNs unaffected by tumor than in corresponding metastatic tumor LNs. In vitro stimulation of primary human T cells in which CD5 was deleted using CRISPR-Cas9 showed that CD5 expression was essential for T cell activation.

To assess what factors might impact CD5+ DCs presence in response to anti-PD-1 in humans, the researchers looked at the DCs from a patient with inherited PD-1 deficiency. This patient had higher frequencies of CD5+ DCs than controls. This led the researchers to hypothesize a role for IL-6, which is excessively produced in this deficiency. In mice treated with anti-PD-1, higher levels of IL-6 were detected in the tumor, and in vitro experiments showed that the presence of IL-6 during DC differentiation resulted in more CD5+ DCs. High concentrations of IL-6 skewed the development from DC1 toward DC2, while low IL-6 concentrations resulted in increased CD5 on both cDC1 and cDC2.

In conclusion, the data presented here suggest an essential role of CD5+ DCs in antitumor responses, in particular, in response to ICB. Treatments directed at increasing the number of CD5+ DCs may therefore be an attractive approach to increase the response to ICB in patients who do not benefit from this therapy.

Write-up by Maartje Wouters, image by Lauren Hitchings.

Meet the researcher

This week, lead author Eynav Klechevsky answered our questions.

From left to right: Mingyu He, Eynav Klechevsky, and Kate Roussak.

What was the most surprising finding of this study for you?
We were surprised to find that CD5 was expressed on mouse DCs. We already knew that it was on human DCs, and finding it in the mouse as well enabled us to develop a mouse model to examine its role in vivo in a preclinical model of disease. By studying the expression and function of CD5 in both species, we gain valuable insights into the role of this protein in the immune system, and how we can leverage this to accelerate the design of more effective treatments for a range of diseases and conditions.

What is the outlook?
As our next step, we aim to investigate how the abundance of CD5+ DCs can serve as a biomarker not only for T cell effector quality, but also for responsiveness to immunotherapy. We aim to understand how the CD5 on DCs facilitate communication with T cells, antigen handling, and directing specific types of T cells, particularly memory T cells, which are crucial in preventing tumor recurrence in patients. In addition, we aim to develop methods to expand these cells in patients and activate them effectively in the context of novel immunotherapy approaches. 

What was the coolest thing you’ve learned (about) recently outside of work?
As a mother and a scientist, I learned that our children get inspired by what we do in the lab. Like me, they are curious about how life works and are enjoying exploring their interests to make exciting discoveries.

References:

He M, Roussak K, Ma F, Borcherding N, Garin V, White M, Schutt C, Jensen TI, Zhao Y, Iberg CA, Shah K, Bhatia H, Korenfeld D, Dinkel S, Gray J, Ulezko Antonova A, Ferris S, Donermeyer D, Lindestam Arlehamn C, Gubin MM, Luo J, Gorvel L, Pellegrini M, Sette A, Tung T, Bak R, Modlin RL, Fields RC, Schreiber RD, Allen PM, Klechevsky E. CD5 expression by dendritic cells directs T cell immunity and sustains immunotherapy responses. Science. 2023 Feb 17.

In the Spotlight...

Intratumoral CD8+ T cells with a tissue-resident memory phenotype mediate local immunity and immune checkpoint responses in breast cancer

Virassamy et al. demonstrated the presence of two phenotypically and transcriptionally distinct CD8+ T cell sub-populations in syngeneic murine TNBC: terminally exhausted CD69+CD103- T cells (TEX) and tissue-resident memory-like CD69+CD103+ T cells (TRM) that transcriptionally resembled TRM cells from healthy murine tissues and human TNBC. Dual-ICB therapy resulted in TCR-Vβ clonal expansion and diversification, increased cytokine production and cytotoxicity of TRM cells in TNBC tumors, and local immune protection against tumor rechallenge by these TRM cells. The CD8+ TRM cell gene signature from tumor-free mice after dual-ICB was associated with better response to ICB in TNBC patients.

Contributed by Shishir Pant

Reducing affinity as a strategy to boost immunomodulatory antibody agonism

Yu et al. examined the impact of affinity on the activity of immunomodulatory antibodies, and showed that lower affinities delivered greater agonism through increased FcγR-independent receptor clustering. A low-affinity anti-CD40 monoclonal antibody exhibited stronger agonistic activity and more effective antitumor activity in vivo compared with the high-affinity parent antibody. More potent agonism mediated by lower affinity could also be recapitulated for a human anti-CD40 antibody. Lowering the affinity converted an inert therapeutic anti-4-1BB antibody (utomilumab) and an antagonistic therapeutic anti-PD-1 antibody (nivolumab) into agonists.

Contributed by Shishir Pant

DSP-0509, a systemically available TLR7 agonist, exhibits combination effect with immune checkpoint blockade by activating anti-tumor immune effects

Ota et al. identified DSP-0509, a structurally unique TLR7-selective agonist suited for i.v. therapy, which activated BMDCs and induced inflammatory cytokines. DSP-0509 inhibited growth of murine syngeneic primary tumors and metastases, particularly if CD8+ TILs were present before treatment. Combining DSP-0509 with anti-PD-1 or anti-CTLA-4 antibodies enhanced antitumor effects, activated TILs, expanded TEM cells, and promoted immune memory. TME RNA analysis showed that DSP-0509 + anti-PD-1 activated T cell and antigen presentation activities and enhanced CTL tumor infiltration and immune effects compared to anti-PD-1 alone.

Contributed by Paula Hochman

Reprogramming of IL-12 secretion in the PDCD1 locus improves the anti-tumor activity of NY-ESO-1 TCR-T cells

To enable TCR activation-dependent cytokine signaling and improve therapeutic T cell functionality without systemic toxicity, Kim et al. modified NY-ESO-1 TCR-transgenic T cells with CRISPR/Cas9 for site-specific insertion of IL-12, replacing PD-1, in the PDCD1 locus. The engineered cells demonstrated antigen-specific IL-12 secretion, loss of PD-1, and tighter control of gene expression than with an NFAT promoter. The IL-12 knock-in improved in vitro T cell cytotoxicity and proliferative capacity after repetitive antigen stimulation, as well as in vivo tumor infiltration and efficacy, over NY-ESO-1 T cell controls or with those with PD-1 knockout alone.

Contributed by Alex Najibi

Overcoming cancer-associated fibroblast-induced immunosuppression by anti-interleukin-6 receptor antibody

Nishiwaki et al. focused on mechanisms underlying the immunosuppressive effects of cancer-associated fibroblasts (CAFs) in the TME. In patient samples of esophageal cancer, high IL-6 levels correlated with significantly shorter OS, tumor immunosuppression, and increased numbers of αSMA+CAFs and immunosuppressive TAMs. In syngeneic tumor models, MR16-1 (anti-IL-6R Ab) suppressed tumor growth, increased CD8+ T cells, and decreased HIF-1α+ expression and activation. Mechanistically, CAF-produced IL-6 increased HIF1α expression, creating competition for glucose in the TME via activation of GLUT-1 and upregulation of VEGF.

Contributed by Katherine Turner

Tregs constrain CD8 + T cell priming required for curative intratumorally anchored anti-4-1BB immunotherapy

To enhance the effectiveness and safety of stimulatory receptor agonist antibodies, Palmeri et al. developed a collagen-targeting 4-1BB agonist Ab (4-1BB-LAIR) and delivered it directly into B16 melanoma tumors along with systemic delivery of a tumor antigen-targeted antibody (TA99). Modest antitumor activity of 4-1BB-LAIR was significantly enhanced by depletion of CD4+ cells, but development of protective memory required depletion of Tregs (FoxP3-DTR or anti-CTLA-4 Ab) and maintenance of effector CD4+ T cells. Activity required a single round of priming in tumor-draining lymph nodes, and 4-1BB-LAIR/TA99 enhanced effector differentiation in the tumor.

Contributed by Ed Fritsch

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.