Weekly Digests
‹ Back to April

Neutrophils help to clear including antigen escape variants in tumors

April 5, 2023

In a previous study, the combination of cyclophosphamide (CTX), CD4+ T cells specific for the melanoma antigen Trp1 (Trp1 T cells), and an anti-OX40 agonist antibody cleared advanced mouse melanoma tumors, including tumor cells that did not express the Trp1 target antigen. Investigating this interesting phenomenon, Hirschhorn et al. found that while T cells were responsible for early antigen-specific killing of tumor cells, complete tumor eradication was dependent on the infiltration and activation of a distinct subset of neutrophils; their results were recently published in Cell.

To begin, Hirschhorn et al. analyzed publicly available patient data, where melanomas were heterogeneous and expressed different combinations and levels of differentiation antigens, including TYRP1, both within and between patients. To study tumor heterogeneity in mice, the researchers utilized two tumor cell lines: B16 melanomas expressing Trp1 (a homolog for human TYRP1), and B78H1 melanomas, which do not express Trp1, nor MHC-I or MHC-II. Triple combination therapy with CTX, Trp1 T cells, and agonist anti-OX40 effectively cleared B16 tumors from mice bearing B16 tumors only, or bearing B16 and B78H1 melanomas on opposite flanks. B78H1 tumors, on the other hand, were not cleared in either setting. However, large heterogeneous tumors consisting of mixed B16 and B78H1 cells were fully eliminated by combination therapy. Replacing the anti-OX40 with anti-CTLA-4 showed similar effects, but was less effective at clearing tumors.

Next, the researchers used bioluminescent imaging to track Trp1 T cells in B16 tumors, and found that peak infiltration did not correlate with maximal tumor regression. Interestingly, while Trp1 T cells were found to be required at early time points (day 3), antitumor effects were maintained when Trp1 T cells were depleted at later time point (day 13), implicating other immune cells in tumor elimination. In RAG-/- mice, which lack adaptive immune cells, similar results were observed, suggesting that the immune cells involved in tumor elimination are a part of the innate immune system.

Quantification of cytokines from tumor extracts showed a strong innate immune signature in treated mice, and further investigation revealed the presence of neutrophils, which were undergoing NETosis – an indicator of maturity and activation. In vitro exposure of naive neutrophils to tumor extracts from mice that had received triple combination treatment induced NETosis and enhanced the neutrophils’ capacity to kill target B78H1 cells. Trp1 cells co-cultured with B16 cells were also capable of educating neutrophils in a similar manner, suggesting that Trp1 T cells secrete factors that promote neutrophil reprogramming.

Investigating the role of neutrophils in vivo using antibody-mediated cell depletion and WT or RAG-/- mice, the researchers showed that neutrophils were required for the efficacy of triple combination treatment in clearing heterogeneous tumors, while other innate immune cells, including monocytes, macrophages, and NK cells, were not. Further, while untreated tumors contained mostly monocytes, macrophages, and DCs, treated tumors contained mostly neutrophils, and exhibited a distinct transcriptional signature.

Parsing out the role of anti-OX40 by replacing it with IgG control in the triple combination therapy, the researchers found in a clustering analysis that about half of cells in the IgG-treated mice were enriched in cluster 0, which showed evidence of an immature phenotype. Meanwhile, the same proportion of cells in the anti-OX40-treated mice were enriched in cluster 1, which showed evidence of activation, chemotaxis, neutrophil function, neutrophil killing, apoptosis, response to IL-1β, NETosis, nitric oxide (NO) biosynthesis, and maturity. Differentiation trajectory analysis showed that clusters 0 and 1 were likely divergent trajectories derived from a common progenitor, with OX40 agonism driving divergent differentiation. When patient data from TCGA was stratified based on signatures of clusters 0 or 1, those with the cluster 1 signature showed favorable survival outcomes.

Further evaluating the effects of OX40 agonism on neutrophils in tumors, the researchers found that exposure to triple combination including anti-OX40 induced higher expression of activation makers, downregulation of CD62L and CD206 (also indicative of increased activation), and reduced expression of PD-L1 compared to triple combination with the control IgG. Additionally, fewer CXCR2+CXCR4- cells were present, indicative of a more mature neutrophil population, and mature (SiglecF+) neutrophils showed higher expression of inducible nitric oxide synthase (iNOS), which could indicate enhanced cytolytic potential via production of reactive oxygen species (ROS).

To investigate the cytolytic activity of neutrophils in tumors following triple combination therapy, Hirschhorn et al. implanted heterogeneous tumors into iNOS-deficient mice. In this setting, triple combination therapy was substantially less effective, and survival was reduced. Ex vivo, neutrophils from this model were less effective at lysing tumor cells, indicating that iNOS and the production of ROS contributes to the antitumor effects of neutrophils. Interestingly, in mice that were deficient in enzymes required for NETosis, the ability of neutrophils to kill tumor cells was unaffected, indicating that while NETosis was an indicator of neutrophil activation, it did not play a direct role in the killing of escape variants.

Analyzing NET formation in patient data, the researchers found that after treatment with neoadjuvant anti-OX40 agonist, tumors from 12 out of 14 evaluated patients showed increased NET formation, with 6 patients showing a more than 2-fold increase. In a mice bearing MC38 colon carcinoma tumors, which respond to agonist anti-OX40, treatment efficacy was reduced when neutrophils were depleted with anti-Ly6G, suggesting that neutrophils play a role in anti-OX40 monotherapy, even in the absence of adoptively transferred antigen-specific T cell therapy. Similar results were observed in data from patients with melanoma following treatment with anti-CTLA-4 and/or anti-PD-1, and again, the depletion of neutrophils in MC38-bearing mice reduced the efficacy of combined checkpoint blockade. When the researchers applied the neutrophil gene signature to a cohort of patients with melanoma who had been treated with anti-CTLA-4 and/or anti-PD-1, they found that the cluster 1 gene signature derived from combination treatment including anti-OX40 was associated with improved outcomes to anti-CTLA-4, while the cluster 0 gene signature showed no difference in expression between groups.

Together, these results show that while antigen-specific T cells play a critical role in early antitumor immune responses, they also secrete factors that recruit and reprogram neutrophils, which help to eliminate antigen escape variants in an iNOS-dependent manner. These effects were shown to be enhanced with OX40 agonism, but also played a role in several other T cell-targeted immunotherapies, including checkpoint blockade, warranting further investigation into the roles of neutrophils and the potential for targeting or utilizing them in immunotherapies.

Write up and image by Lauren Hitchings

References:

Hirschhorn D, Budhu S, Kraehenbuehl L, Gigoux M, Schröder D, Chow A, Ricca JM, Gasmi B, De Henau O, Mangarin LMB, Li Y, Hamadene L, Flamar AL, Choi H, Cortez CA, Liu C, Holland A, Schad S, Schulze I, Betof Warner A, Hollmann TJ, Arora A, Panageas KS, Rizzuto GA, Duhen R, Weinberg AD, Spencer CN, Ng D, He XY, Albrengues J, Redmond D, Egeblad M, Wolchok JD, Merghoub T. T cell immunotherapies engage neutrophils to eliminate tumor antigen escape variants. Cell. 2023 Mar 30.

In the Spotlight...

Lineage tracing reveals clonal progenitors and long-term persistence of tumor-specific T cells during immune checkpoint blockade

Pai et al. performed scRNA/TCRseq on T cells from three patients with non-small cell lung cancer (31 tissue regions, including tumors, adjacent normal tissues, and lymph nodes) to systematically profile the regional distribution, region-dependent cell states, and long-term persistence of tumor-specific T cells post ICB. Clonally expanded CD8+ T cells, Tregs, and follicular helper CD4+ T cells with tumor-specific features showed progressive exhaustion in proximity to viable cancer cells, and were clonally linked with TCF7+ progenitor exhausted T cells in lymph nodes. Tumor-specific T cell clones persisted in the blood for years after ICB therapy.

Contributed by Shishir Pant

An immunometabolism subtyping system identifies S100A9+ macrophage as an immune therapeutic target in colorectal cancer based on multiomics analysis

Bao et al. integrated transcriptome, proteome, and single-cell transcriptome analysis to develop immunometabolism molecular subtyping (IMS), which subdivided patient CRC into three subtypes (C1, C2, and C3) with distinct metabolic and immunogenic properties, and associated clinical responses. The C3 subtype exhibited the worst prognosis, had a high abundance of immunosuppressive macrophages, and was enriched for S100A9+ macrophages/monocytes compared to C1 and C2 subtypes. Tasquinimod (an S100A9 inhibitor) and anti-PD-1 dual treatment reprogrammed the pro-tumor M2-type TAMs into antitumor M1-type TAMs and induced a strong antitumor immune response.

Contributed by Shishir Pant

IFNγ-induction of TH1-like regulatory T cells controls antiviral responses

To understand the drivers of Treg immunosuppression, Gocher-Demske et al. used LCMV and influenza infection models and genetic knockouts to show that IFNγ, derived mostly from CD8+ T cells, stimulated a stable TH1 program in Tregs, which subsequently limited CD8+ T cell inflammation. TH1 Tregs were highly immunosuppressive toward antigen-specific CD8+ T cells, reduced CD8+ memory cell formation, and limited the magnitude of CD8+ T cell vaccine responses. Treg-specific absence of the IFNγ receptor resulted in less suppressive TH2 Tregs, with concomitant weight loss and some signs of tissue pathology. IL-12 had little effect on the TH phenotype in the viral models.

Contributed by Ed Fritsch

Inflammatory CD4/CD8 double-positive human T cells arise from reactive CD8 T cells and are sufficient to mediate GVHD pathology

Focusing on identifying T cells responsible for GVHD pathology, Hess et al. analyzed a cohort of 35 patients who received allogeneic hematopoietic cell transplants and showed that CD4+/CD8+ double-positive T cells (DPT) correlated with ≥ grade 2 GVHD, but were absent in the initial graft. In a xenogeneic transplant model, DPT cells arose from antigen-stimulated CD8+ T cells and were transcriptionally, metabolically, and phenotypically distinct from single-positive CD4+ and CD8+ T cells. Isolated DPTs were sufficient to drive xeno-GVHD pathology without providing a survival benefit when transplanted into naive mice, suggesting DPTs may play a direct role in GVHD pathology.

Contributed by Katherine Turner

Neutralizing IL-8 potentiates immune checkpoint blockade efficacy for glioma

Liu et al. observed that the majority of T cells in high- and low-grade gliomas of 152 patients were in the perivascular cuffs. TIL/PBMC comparison showed elevated TIL expression of CXCR3, CCR5, CD25 and PD-1, and identified CD8+ and CD4+ TEFF TILs, pre-/exhausted CD8+ TILs, and a terminally differentiated, non-clonal IL-8+ CD4+ TIL population with innate-like features, which was associated with poor patient outcomes. In IL-8-humanized and cell transfer mouse models, IL-8-producing CD4+ T cells, myeloid cells, and tumor cells induced MDSC infiltration and angiogenesis, which promoted tumor growth and restricted ICB. Anti-PD-1 response was bolstered by interdiction of the IL-8/CXCR1/2 pathway.

Contributed by Paula Hochman

The molecular and functional landscape of resistance to immune checkpoint blockade in melanoma

Lim and Shklovskaya et al. probed mechanisms of ICB resistance in paired tumor biopsies and cell lines from 18 patients with melanoma who were resistant to checkpoint therapy. The cell lines were highly heterogeneous: one was IFNγ unresponsive, while others had intrinsic IFNγ signaling, all with low immune infiltration. Half acquired a “de-differentiated” phenotype with loss of melanocyte antigens (e.g., Melan-A) and few activated and responsive T cells in biopsies and in vitro assays, respectively. Alterations in MHC-I/II (through CTIIA or B2M loss or epigenetic changes) were also common, and several lines, primarily from brain metastases, had PTEN deficiency.

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.