Weekly Digests
‹ Back to November

Activating macrophages to remodel the tumor microenvironment

November 29, 2023

One of the reasons immune checkpoint blockade (ICB) may fail in solid tumors like pancreatic ductal adenocarcinoma (PDA) is a lack of T cell infiltration and extensive myeloid inflammation in the tumor microenvironment (TME). Wattenberg et al. aimed to overcome these characteristics in recent work published in Science Immunology.

Given the known plasticity of myeloid cells, the researchers started out by assessing publicly available scRNAseq data of human PDA to look for the expression of putative myeloid activating receptors. One of the most abundantly expressed TNF receptor family members was CD40. While strategies to target CD40 in early clinical studies have had limited effects, the researchers hypothesized that combining it with another agent targeting a myeloid signaling pathway might be more effective in triggering antitumor immune responses. The myeloid cells also expressed pattern recognition receptors (PRRs), frequently on clusters that lacked robust CD40 expression, suggesting these might be rational targets for combination therapy.

To test this hypothesis, the researchers made use of ICB-resistant GEMM PDA mouse models. As in the clinic, treatment with an agonistic CD40 antibody (agCD40) had a limited effect on tumor control. However, combining this therapy with any of several agonists of various PRRs resulted in high cure rates. The researchers focused subsequent work on targeting Dectin-1, which is a receptor for β-glucans (BG), because BG can be systemically administered in humans without causing significant toxicity. Combined treatment of BG and agCD40 in PDA models induced tumor control, without overt toxicity.

The researchers then performed mass cytometry (CyTOF) on orthotopic tumors to assess the underlying mechanisms of this synergy. A broad remodeling of the myeloid and lymphoid populations in the tumor was observed post-treatment. In the myeloid compartment, BG monotherapy increased the frequencies of PD-L1-Ly6G+ granulocytes, and a shift toward CD206loF4/80+ macrophages (CD206 is a marker of immunosuppressive macrophages) was observed. Meanwhile, agCD40 monotherapy increased inflammatory monocytes and IA/IE+F4/80+ macrophages. Combined treatment also induced these effects, and decreased immunosuppressive CD206+PD-L1+ macrophages. In the T cell compartment, agCD40 increased the frequency of Ki67+ICOS+CD127+CD4+ T cells and CD8+ T cells with low expression of immune checkpoints, while it decreased the frequency of Tregs. Bulk RNAseq of the treated tumors showed that BG resulted in the enrichment of genes related to innate immune activation, cytokine production, myeloid migration pathways, and myeloid-recruiting chemokine expression, while agCD40 was associated with increases in gene pathways related to lymphocyte and T cell activation.

To assess whether T cells were required for the effects of BG/agCD40, CD4+ and CD8+ T cells were depleted. Depleting both T cell subsets abolished the effects of therapy, and depleting one of the two populations resulted in a partial decrease in antitumor effects, indicating both populations were important. In cured mice, a second tumor challenge showed that treatment induced T cell-dependent immunological memory. Looking at the importance of other immune cell subsets, the researchers found that cDC1 were essential for the effects of the combined therapy, as in Batf3ko mice, deficient in cDC1, the antitumor effects were diminished. These data suggest that both T cells and cDC1 were required for the antitumor effects of BG/agCD40.

IHC analysis showed that agCD40 drove the increased tumoral infiltration of T cells. However, the infiltrated T cells in the TME after treatment were present surrounding the tumor lesions, not in the lesions. In mice lacking MHC class I and II or in Prf1ko mice, which lack perforin, the combination treatment remained effective. These data suggest that even though the T cells were required, they did not function as cytotoxic effectors in this setting.

Looking more in depth at the intratumoral T cells using scRNAseq data at 8 days after treatment, the researchers confirmed that the major changes in the lymphoid compartment were due to T cells and were driven by agCD40 treatment, with decreases in Tregs and increases in CD4+Tcf7+ and CD4+Tox+ clusters. There was also an increase in the frequency of CD8+Cxcr6+ T cells, a stem-like population, and CD8+Cx3cr1+ T cells, a tumor-specific effector memory-like population. Genes related to positive regulation of cytokine production were highly expressed in T cell subsets after agCD40 or combination therapy. Ifng was one of the highest expressed cytokines by T cells in combination treatment groups. Intratumoral IFNγ administration in the absence of agCD40 could also trigger the antitumor activity of BG therapy and increase survival in PDA models. These data suggest that agCD40 results in T cell infiltration and IFNγ production, which, combined with BG, mediates the antitumor effects.

Diving into the effects exerted by BG, the researchers assessed the Dectin-1-dependent binding of BG, and found that at 18 hours post-treatment, it mostly bound to Ly6Chi monocytes and Ly6G+ granulocytes, but not to TAMs, T cells, B cells, or CD45- cells. Tumors from treated mice showed an increase in the TAM-to-tumor cell ratios, suggesting that BG may remodel the TME and affect TAMs.

TAMs were common in the stromal compartment of the TME, and combination treatment increased intratumoral TAMs. scRNAseq data identified four TAM and two monocyte populations in the TME. CD206+ TAMs, previously associated with T cell exhaustion and protumoral characteristics, were found in the tumor border region, while CD206- TAMs were found in the intratumoral region. BG/aCD40 therapy resulted in a decrease in the ratio of CD206+/CD206- TAMs. TAMs showed increased immunostimulatory transcriptional programs, including defense response and biotic stimulus (stimulus from a component of a living organism), and CD206- TAMs had increased peroxisome activity after combination treatment. BG was a major driver of these changes, but upregulation of the IFNγ-responsive genes Stat1 and Ly6a was associated with agCD40 treatment, and pSTAT1tyr701+ TAMs could be detected in the TME, suggesting both treatments impacted the TAM populations.

Finally, to confirm the role of TAMs in treatment responses, BG/agCD40 treatment was repeated in combination with an anti-Ly6C antibody to deplete Ly6C+ cells, which showed reduced efficacy of the treatment. Additionally, pharmacologic targeting of CSF1-CSF1R signaling to inhibit systemic phagocytic cells and intratumoral TAMs also reduced efficacy.

In conclusion, this research shows the benefits of combining PRR stimulation (BG) and agCD40 treatment in preclinical ICB-resistant models of PDA, resulting in a profound remodeling of the TME and activation of immunostimulatory TAMs. This provides new avenues for clinical research in patients with PDA and other ICB-resistant cancer types.

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

Meet the researcher

This week, first author Max M. Wattenberg answered our questions.

First author Max M. Wattenberg

What was the most surprising finding?
One of the most surprising findings in our study was that cytotoxic T cells did not need to directly engage tumor cells for effective immunosurveillance. Rather, combinatory myeloid cell activating therapy drove T cells in the stroma to secrete immunostimulatory cytokines, which educated antitumor effector myeloid cells. These findings suggest new ways in which antigen-specific T cell responses might be leveraged to treat cancer. Indeed, our data show that by targeting myeloid cells, a concerted innate and adaptive immune response to cancer can be generated, bypassing classical mechanisms of immune evasion, including T cell exclusion and exhaustion.

What is next?
There are two key next steps for this work. First, we plan to define the safety, efficacy, and immunological mechanisms of combined myeloid activation in humans. To address this, we have initiated a Phase 1b clinical trial studying combined CD40 and Dectin-1 activation for the treatment of patients with pancreatic cancer (NCT05484011). Second, we aim to determine the molecular mechanisms by which myeloid activating therapy is mediating tumor cell killing, and in doing so, provide a roadmap for the development of next-generation myeloid-activating therapies.

What is the coolest thing you have learned outside of work recently?
I recently attended a college basketball game at The Palestra on the University of Pennsylvania’s campus, and was fortunate to see the Quakers upset nationally ranked Villanova. The Palestra, known as “The Cathedral of College Basketball”, opened in 1927, and hosted the East Regional of the NCAA’s first national tournament in 1939. Further, The Palestra has witnessed more college basketball games than any other arena, and is one of the first steel-and-concrete arenas in the United States.

References:

Wattenberg MM, Coho H, Herrera VM, Graham K, Stone ML, Xue Y, Chang RB, Cassella C, Liu M, Choi-Bose S, Thomas SK, Choi H, Li Y, Markowitz K, Melendez L, Gianonne M, Bose N, Beatty GL. Cancer immunotherapy via synergistic coactivation of myeloid receptors CD40 and Dectin-1. Sci Immunol. 2023 Nov 17. 

In the Spotlight...

Association between pretreatment emotional distress and neoadjuvant immune checkpoint blockade response in melanoma

Fraterman and Reijers et al. evaluated the association between pretreatment emotional distress (ED) and clinical response in stage III melanoma patients treated with neoadjuvant ICB in phase II PRADO trial. Patients with pretreatment ED showed a reduced MPR (46% versus 65%) and significantly impaired 2-year RFS (74% versus 91%) and 2-year DMFS (78% versus 95%) compared with patients without ED, after adjusting for the previously identified baseline biomarkers (IFNγ signature and TMB). Transcriptional analyses of baseline β-Adrenergic- or glucocorticoid-associated pathways did not show significant association with ED.

Contributed by Shishir Pant

NMDAR antagonists suppress tumor progression by regulating tumor-associated macrophages

Yuan, Hu, and Ju et al. showed that in vitro blockade of the ion channel N-methyl-D-aspartate receptor (NMDAR) polarized macrophages toward an M1-like phenotype. NMDAR antagonist treatment of mice bearing fibrosarcoma or liver tumors decreased immunosuppression by TAMs, boosted TIL effector function, inhibited tumor growth, and prolonged host survival. scRNAseq showed NMDAR regulated Ca++ influx, ROS production, and mitochondrial function in macrophages, and decreased immunosuppression by TAMs to promote T and NK cell antitumor activity. NMDAR antagonism improved anti-PD-1 antibody efficacy against liver tumors established in mice.

Contributed by Paula Hochman

DGKα/ζ inhibition lowers the TCR affinity threshold and potentiates antitumor immunity

Kureshi et al. demonstrated that dual DGKα/ζ inhibition enhanced priming and antigen-specific effector functions of CD8+ T cells with different affinities (TRP1high and TRP1low) in vitro and in vivo. The effect was most pronounced in situations where TCR affinity or avidity were suboptimal. DGKi-mediated cytotoxicity was dependent on MHC-I and IFNγ expression, but not on perforin. DGKi increased proliferation and cytokine production in antigen-specific T cells in mice bearing C2VTrp1 tumors (low Trp1 expression) and synergized with anti-PD-1 therapy to control melanoma and pancreatic tumor growth in an antigen-dependent manner.

Contributed by Shishir Pant

An engineered immunocytokine with collagen affinity improves the tumor bioavailability, tolerability, and therapeutic efficacy of IL-2

Silver et al. fused vWF collagen binding domain (CBD) to F10 – a fusion comprising human IL-2 and an anti-IL-2 Ab that blocks IL-2/IL-2Rα and boosts IL-2/IL-2Rβ binding to bias TEFF over Treg cell activation – to create F10 IC-CBD. Compared to controls, intratumoral (i.t.) injection of F10 IC-CBD exhibited increased/prolonged bioavailability and bioactivity in situ, minimal systemic exposure, a wider safety margin, and enhanced/potent efficacy in tumor-bearing mice. This was due to collagen binding and TEFF bias. I.t. injection of F10 IC-CBD also elicited systemic immunity, as evidenced by an abscopal response, and its antitumor efficacy was potentiated by systemic PD-1 blockade.

Contributed by Paula Hochman

In situ vaccination via tissue-targeted cDC1 expansion enhances the immunogenicity of chemoradiation and immunotherapy

Fusion of Flt3L to albumin (Alb-Flt3L) extended the half-life of Flt3L and improved its lymph node and tumor accumulation. I.v. Alb-Flt3L expanded cDC1s and pDCs in mouse spleens, tumors, and tumor-draining lymph nodes, and activated intratumoral T cells. Combined with radiation therapy, Alb-Flt3L induced tumor neoantigen-specific T cells and controlled TC-1 and MC38 tumors, and was superior to Flt3L. Alb-Flt3L also synergized with cisplatin chemotherapy to treat large tumors, dependent on Batf3 and type-I IFN. Efficacy was further improved with anti-PD-L1. In CD34-humanized mice, Alb-Flt3L expanded human cDCs and pDCs more so than Flt3L.

Contributed by Alex Najibi

Prior antibiotic administration disrupts anti-PD-1 responses in advanced gastric cancer by altering the gut microbiome and systemic immune response

Kim, Koh, S-J Shin and J-H Shin et al. addressed the impact of prior antibiotic (pATB) administration in patients with advanced gastric cancer following PD-1 blockade. In multiple patient cohorts, pATB significantly decreased progression-free survival and overall survival, whereas outcomes in a patient cohort treated with chemotherapy (irinotecan) were unaffected by pATB administration. Mechanistically, pATBs significantly decreased gut microbiome diversity, reduced numbers of Lactobacillus gasseri, and disproportionately enriched circulating exhausted CD8+ T cells levels, all of which have been implicated in worse outcomes and decreased responses to PD-1 blockade.

Contributed by Katherine Turner

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.