Weekly Digests
‹ Back to February

TLR agonism and PD-1 blockade unfortunately activate suppressor cells

February 26, 2025

The activation of an immune response involves a system of checks and balances. While immunotherapies can be used to tip the scales, they are not always effective, as resistance mechanisms may act to counter desired responses. In research recently published in Science Translational Medicine, Nishinakamura and Shinya et al. found that a combination of the TLR agonist OK-432 plus PD-1 blockade failed to induce antitumor responses in immune-cold tumor models. Upon further investigation, they were able to attribute this resistance to an unintended co-activation of PMN-MDSCs, which could be overcome through PMN-MDSC depletion.

To begin, Nishinakamura and Shinya et al. evaluated OK-432 – an agonist for multiple TLRs – against human DCs derived from both healthy donors and patients with gastrointestinal cancers, and found that it effectively induced DC maturation, enhancing expression of CD86, CD80, CD40, and PD-L1. Functionally, OK-432 also improved DC priming of peptide-specific CD8+ T cells, and this effect was further enhanced with the addition of anti-PD-1.

Moving into mouse models, the researchers investigated the combination of OK-432 and anti-PD-1 in a variety of cold tumor models, including LL/2 Lewis lung carcinoma, CT26 colon carcinoma, and CT26-NY-ESO-1. While the combination treatment slightly inhibited tumor growth in the CT26 model and slightly prolonged survival in the CT26-NY-ESO-1 model, there was no advantage over either monotherapy, and no advantage at all in the LL/2 model.

Investigating this on an immunological level, the researchers evaluated tumors by flow cytometry and found that while OK-432 effectively upregulated CD80 and PD-L1 on DCs, CD8+ T cells, CD4+ T cells, and Tregs were all unexpectedly reduced in frequency. Bulk RNAseq, gene set enrichment analysis, and CyTOF/UMAP analyses each suggested that MDSCs, and particularly PMN-MDSCs, were upregulated in tumors. These results were further confirmed experimentally, with flow cytometry showing an increase in Ly6G+ PMN-MDSCs, but not Ly6C+Ly6G- M-MDSCs in tumors upon treatment with OK-432. Further, CD8+ T cells cocultured with FACS-purified PMN-MDSCs showed reduced proliferation, reduced production of IFNγ, and limited capacity for cell killing.

Looking deeper into the mechanism by which PMN-MDSCs accumulated in tumors, the researchers found that Ly6G+ cells were reduced in the peripheral blood and bone marrow of mice 24 hours after OK-432 administration, suggesting that PMN-MDSCs likely migrated from these locations to tumors upon with treatment OK-432. Evaluating cytokines and chemokines that might mediate this migration, the researchers noted upregulation of IL-6 and CXCL1 in tumors. Evaluation of IL-6 via analysis of STAT3 activation ruled out a role for IL-6 in the recruitment of PMN-MDSCs. However, the increase in CXCL1 production was found to originate from peritoneal macrophages (but not tumor cells) in the TME and was dependent on TLR–MyD88–NFκB signaling. Further, CD45+CXCR2+ cells in the TME highly expressed Ly6G after OK-432 treatment, suggesting that PMN-MDSCs were likely recruited via the CXCR2–CXCL1 axis.

To determine how different tumors might respond to OK-432, Nishinakamura and Shinya et al. investigated its effects on 10 different cancer cell lines. Bulk RNA sequencing along with UMAP and clustering analysis were performed, and different tumor cell lines were grouped based on their haplotypes and levels of immune infiltration. While the top three most highly immune-infiltrated lines (MC38, MC38-OVA, and EMT6) responded to a combination of Ly6G+ cell depletion (using anti-Ly6G) and anti-PD-1, the remaining cell lines, including LL/2, CT26, and CT26NY-ESO-1, did not. However, when the researchers tested a triple combination of OK-432 plus anti-PD-1 plus Ly6G+ cell depletion (using anti-Ly6G or CXCR2-neutralizing antibodies), treatment effectively inhibited tumor growth and prolonged survival compared to controls, monotherapies, and double combination therapies in immune-cold models.

Looking within the PMN-MDSC subset induced by OK-432, the researchers found that in CT26-NY-ESO-1 tumors, treatment induced a particularly suppressive subset of CD14high PMN-MDSCs. However, triple combination therapy with the anti-Ly6G reduced these Ly6G+CD14high PMN-MDSCs, along with Ly6G+CD14intermed and Ly6G+CD14- PMN-MDSCs. Use of CXCR2-neutralizing antibodies was not as effective as anti-Ly6G in the triple combination, which may be due to differences in the potency of the antibodies towards this highly suppressive subset. Similar results were observed in an LL/2 tumor model. Further, CD8+ T cells, but not CD4+ T cells or NK cells in the LL/2 tumor model were required for the antitumor efficacy of triple combination treatment. Triple combination treatment also increased antigen-specific CD8+ T cells and their effector functions (granzyme B and IFNγ production) compared to OK-432 or anti-Ly6G alone. A CXCR2 inhibitor could also be used in place of anti-Ly6G or CXCR2-neutralizing antibodies as a means of inhibiting PMN-MDSCs, showing similar antitumor effects when used in triple combination with OK-432 and anti-PD-1.

Finally, Nishinakamura and Shinya et al. investigated whether their results in mice were relevant in patients by evaluating patients with lung cancer who were treated with OK-432 to control pleural effusion, as it is approved for this use in Japan. In these patients, concentrations of CXCL1 were increased in the pleural effusion after treatment, as were levels of CD14+ cells among CXCR2+CD45+ cells, and expression of LOX1 (a marker of human PMN-MDSCs) on a per cell basis, which aligns with an increase in PMN-MDSCs in response to treatment.

Overall, these results suggest that while TLR agonism may activate DCs and enhance their capacity for T cell priming, it also recruits immunosuppressive PMN-MDSCs, limiting efficacy alone or in combination with anti-PD-1 in immune-cold tumor models. However, depletion of PMN-MDSCs could be used to restore the efficacy of TLR agonism plus anti-PD-1, shifting the balance towards immune activation and antitumor efficacy.

Write-up and image by Lauren Hitchings

References:

Nishinakamura H, Shinya S, Irie T, Sakihama S, Naito T, Watanabe K, Sugiyama D, Tamiya M, Yoshida T, Hase T, Yoshida T, Karube K, Koyama S, Nishikawa H. Coactivation of innate immune suppressive cells induces acquired resistance against combined TLR agonism and PD-1 blockade. Sci Transl Med. 2025 Feb 12.

In the Spotlight...

CV1-secreting sCAR-T cells potentiate the abscopal effect of microwave ablation in heterogeneous tumors

Cao, Liu, and Xiao et al. examined the effect of combining microwave ablation (MWA) with sCAR T cells secreting CV1 (a short lived, affinity-enhanced SIRPα that blocks CD47 signaling) on the abscopal effect in antigen-heterogeneous tumor models that respond poorly to CAR T cell therapy alone. As in humans, MWA alone elicited a weak abscopal response. However, MWA plus sCAR-T secreting CV1 not only potentiated local antitumor effects, but stimulated chemokine production by ablated tumors, which recruited and activated macrophages, promoted M1 polarization, and enhanced phagocytosis at distant non-ablated tumor sites, without added systemic toxicity.

Contributed by Katherine Turner

EZH1/EZH2 inhibition enhances adoptive T cell immunotherapy against multiple cancer models

Porazzi and Nason et al. showed that EZH2 inhibition sensitizes a broad spectrum of tumors to diverse adoptive T cell immunotherapies. EZH2 inhibition enhanced tumor immunogenicity and modulated inflammation and apoptosis pathways, leading to improved tumor–CAR T cell interactions and antitumor efficacy. EZH2 inhibition/KO in CART19 did not improve tumor killing, nor compromise its antitumor effect. Tumor inhibition of both EZH1 and EZH2 further enhanced CAR T cell activation, expansion, and infiltration, leading to improved antitumor efficacy across diverse liquid and solid cancer models.

Contributed by Shishir Pant

Isolation of a tumor neoantigen specific CD8+ TCR from a skin biopsy of a vaccination site

Roberti et al. found that vaccine site-infiltrating lymphocytes (VILs) isolated from a patient with TNBC who was treated with a personalized neoantigen peptide vaccine included both Trm-like CD4+ and CD8+ T cells with distinct antigen reactivities from PBMCs. One neoantigen (NCOR1L1475R) enabled CD8+ T cell expansion and formed stable HLA-I complexes, and TCR-T cells generated using reactive clonotypes responded to the neoantigen, but not the wild-type peptide. Post-vaccine tumor biopsy showed immune activation, but also loss of HLA-I expression and the NCOR1L1475R mutation, and NCOR1L1475R- reactive T cells were not found in the tumor or blood - only in VILs.

Contributed by Alex Najibi

SARS-CoV-2 B Epitope-Guided Neoantigen NanoVaccines Enhance Tumor-Specific CD4/CD8 T Cell Immunity through B Cell Antigen Presentation

Li et al. engineered SARS-CoV-2 B cell epitope-guided nanovaccines (BSARS) to stimulate BCR crosslinking and B cell-mediated antigen presentation, unlike most current cancer vaccines that depend on DC or macrophage antigen presentation. When compared to traditional neoantigen vaccines, BSARS neoantigen peptide or mRNA nanovaccines displayed improved efficacy in four murine tumor models. In addition to activation of a classical germinal center response, the BSARS nanovaccines increased tumor-specific follicular and non-follicular CD4+ T cells, and induced tumor-specific CD8+ T cells in a way that depended on B cell and CD4+ T cell activation.

Contributed by Ute Burkhardt

Shared pathway of WDFY4-dependent cross-presentation of immune complexes by cDC1 and cDC2

Jo et al. characterized cDC1 (Δ32 mice)- and cDC2 (Δ1+2+3 mice)-deficient mouse models and evaluated the role of DC subsets in the priming of CD4+ and CD8+ T cells in response to several forms of antigen in vivo. cDC1s were necessary and sufficient for the priming of both CD4+ and CD8+ T cells for cell-associated antigens, but dispensable for antigens derived from immune complex (IC) antigens. cDC2 were sufficient for cross-presentation of IC antigens in cDC1-deficient (Δ32 mice), leading to the induction of CD8+ T cell responses that protected against immunogenic tumors in a WDFY4-dependent manner.

Contributed by Shishir Pant

Targeting vaccines to dendritic cells by mimicking the processing and presentation of antigens in xenotransplant rejection

Wang et al. showed that xenogeneic cell membrane-derived vesicles (XMVs) encapsulating defined antigens and coated with antibodies targeting the xenogenic tissue promoted DC activation, antigen cross-presentation, and trafficking to mouse LNs. Injecting mice with XMVs containing tumor-derived antigenic peptides or XMVs coated onto LNPs encapsulating an mRNA coding for a tumor antigen induced specific T cell-mediated inhibition of tumor growth. Mice injected with XMV-coated LNPs encapsulating mRNA encoding the SARS-CoV2 spike protein induced higher titers of antigen-binding and neutralizing Abs than a commercial mRNA–LNP formulation.

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.