Weekly Digests
‹ Back to September

IFNγ signaling paves the way for effective checkpoint blockade

September 16, 2020

In recent years, researchers have made much progress in understanding the therapeutic response to checkpoint blockade strategies, but to further improve the success rate, clues might be found in a deeper investigation of the mechanisms behind the response. In a recent publication in Cancer Cell, Grasso et al. describe their analysis of a large number of prospective biopsy samples from metastatic melanoma patients treated with anti-PD-1 (Nivolumab) or anti-PD-1 combined with anti-CTLA-4 (Ipilimumab) as part of the Checkmate 038 trial. Transcriptome analysis by bulk RNAseq, with modeling and validation of pathways of interest in 58 human melanoma cell lines, could then be correlated with clinical response to uncover these mechanism-based clues.

From 101 patients, 84 baseline and 85 on-therapy samples were subjected to RNAseq. Of these, 68 samples were paired, and for the unpaired samples, an optimal pooled t-test was applied to enable comparisons before and after treatment. Clinical response was defined as complete or partial response (CRPR), while non-responders had stable disease (SD) or progressive disease (PD).

The researchers first analyzed the immune infiltrate using denominated microenvironment cell populations-counter (MCP-counter), which deconstructs RNAseq data to define immune cell types. Patients responding to therapy had higher baseline levels of T cells, B lineage cells, myeloid dendritic cells, and NK cells than non-responders. In general, all patients had increases in T cell infiltration upon therapy, but the changes were most extensive in responders. Principal component analysis confirmed that the most predictive factor was the degree of T cell infiltration in both the baseline and the on-therapy samples. Furthermore, t-SNE embedding to cluster the genes with their closest immune cell signature derived from MCP-counter resulted in two gene clusters; one that was correlated with the T cell score, and one that was anti-correlated, further confirming the importance of T cell infiltrate for therapy response.

Given the clear predictive value of T cell infiltration, the researchers then studied the drivers behind this response by examining effector cytokines and toxic granules associated with T cell activity. They found a higher expression of genes encoding perforin, granzyme B, TRAIL, and TNFα in responders. These expression patterns followed the expression of IFNγ, which led them to hypothesize that cancer cell expression of interferon response genes might be relevant for attracting immune cells and stimulating their function. This suggests that patients with a lack of response should have melanoma cells incapable of responding to IFNγ.

To study this hypothesis, Grasso et al. analyzed the transcriptome, before and after IFNγ treatment, of 57 previously established primary human melanoma cell lines, as well as the non-malignant melanocyte cell line HeMa. Upon IFNγ treatment, changes in IFNγ response genes were found in most cell lines. The responding cell lines had a strong upregulation of genes associated with the antigen processing and presentation machinery, increases in chemokines such as CXCL9, CXCL19, and CXCL11, and other genes in interferon signaling pathways. However, as anticipated, a distinct subset of 12 cell lines, characterized by JAK1 or JAK2 loss-of-function mutations, did not signal through the IFNγ receptor.

The researchers then went back to the biopsies and found that responders had a higher baseline expression of interferon response genes than non-responders, and that the expression further increased upon treatment. Within the group of samples from non-responders, there were even samples without expression of interferon response genes at baseline. The main differences between responders and non-responders were a higher expression in antigen processing and presentation pathway genes in responders, particularly in genes encoding HLA class I and II, B2M, TAP1, TAP2, NLRC5, and CIITA. To correct for the fact that these samples contained all cells in the microenvironment and not just tumor cells, the researchers normalized the data for LCA (CD45). After this correction, the HLA I score was not different in responders, suggesting that this signal was driven by immune cell infiltration. Therefore, these data should be validated by single cell RNAseq to determine the cell type driving these signatures.

The authors then looked at two pathways recently suggested to be associated with immune exclusion in the tumor microenvironment. First, they assessed WNT signaling by using a nine-gene RNAseq-based WNT score. Responders had a significant decrease in the score after therapy, while the score did not change in non-responders. Tracking the WNT score and T cell infiltration between baseline and therapy samples suggested a relatively random distribution in non-responders, while the responders trended from high WNT/low T cell to a low WNT/ high T cell infiltrate between before and on-therapy samples. The same trend of a lower WNT score after treatment with IFNγ was found in the cell lines, as well as downregulation of MYC, a target gene in the WNT signaling pathway.

Second, the researchers analyzed the recently developed ‘Jerby-Arnon’ immune exclusion signature; the main driver of that signature, CDK4; and PAK4, a gene recently found to be overexpressed in patients with low T cell infiltrates who do not respond to anti-PD-1 therapy. The signature and both genes were downregulated in responders – effects that were not found in the IFNγ-treated cell lines – suggesting these are likely downstream effects.

In summary, the results presented by Grasso et al. indicate changes in gene signatures related to immune infiltration in patients responding to checkpoint therapy. Enhanced IFNγ signaling, triggered by the activity of pre-existing T cells in the tumor, resulted in increases in genes related to the antigen processing and presenting pathways, and increases in chemokines, resulting in more immune cell attraction. This was further aided by a reduction in WNT signaling and downstream effects on genes of the ‘Jerby-Arnon’ immune exclusion signature, CDK4, and PAK4. Together, this achieved a more welcoming immune environment in responders.

The mechanisms behind the therapy response to checkpoint inhibition presented here could spark new research into potential combination treatment strategies. Raising interferon signaling in tumor cells using, for instance, oncolytic viruses that trigger IFNγ production or intratumoral TLR stimulation, may pave the way for improved outcomes to checkpoint blockade in patients who currently do not respond to therapy.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, first author Catherine Grasso answered our questions.

First author Catherine Grasso and lead author Antoni Ribas

What prompted you to tackle this research question?
The initial motivation to tackle this problem was the observation that immune infiltration seemed to be a primary driver of response. We wanted to break down the samples and see if there were different responses for different immune cell types.

What was the most surprising finding of this study for you?
We looked at a cell line cohort to identify additional mutations, potentially in the interferon gamma pathway, driving resistance to interferon gamma, and we didn't find them. The ones we knew, B2M and JAK1/2, were present and we did not find more. With that said, we found that immune infiltration before treatment played a large role, and that the mutations or gene expression driving infiltration played a much larger role.

What was the coolest thing you’ve learned (about) recently outside of work?
It's possible to cure schizophrenia using immunotherapy.
https://www.nytimes.com/2018/09/29/opinion/sunday/schizophrenia-psychiatric-disorders-immune-system.html

References:

Catherine S. Grasso, Jennifer Tsoi, Mykola Onyshchenko , Gabriel Abril-Rodriguez, Petra Ross-Macdonald, Megan Wind-Rotolo, Ameya Champhekar, Egmidio Medina, Davis Y. Torrejon, Daniel Sanghoon Shin, Phuong Tran, Yeon Joo Kim, Cristina Puig-Saus, Katie Campbell, Agustin Vega-Crespo, Michael Quist, Christophe Martignier, Jason J. Luke, Jedd D. Wolchok, Douglas B. Johnson, Bartosz Chmielowski, F. Stephen Hodi, Shailender Bhatia, William Sharfman, Walter J. Urba, Craig L. Slingluff, Jr., Adi Diab, John B.A.G. Haanen, Salvador Martin Algarra, Drew M. Pardoll, Valsamo Anagnostou, Suzanne L. Topalian, Victor E. Velculescu, Daniel E. Speiser, Anusha Kalbasi, and Antoni Ribas. Conserved Interferon-g Signaling Drives Clinical Response to Immune Checkpoint Blockade Therapy in Melanoma- Cancer Cell sept 10, 2020

In the Spotlight...

Effective combination immunotherapy using oncolytic viruses to deliver CAR targets to solid tumors

Park et al. selectively infected intractable human solid tumor cells with an oncolytic vaccinia virus (OV19t) encoding a truncated CD19 protein (CD19t) and detected intracellular OV19t and CD19t on tumor cell surfaces prior to OV-mediated tumor lysis. CD19-CAR T cells were activated and secreted IFNγ and IL-2 when co-cultured with OV19-infected tumor cells, killed OV19t-infected tumor cells in vitro, and regressed OV19t-infected tumors in multiple xenograft models. Endogenous and transferred murine (m) CD19-CAR splenic T cells infiltrated and regressed syngeneic s.c. tumors expressing mOV19t-encoded mCD19t.

Contributed by Paula Hochman

A Critical Role of CD40 and CD70 Signaling in Conventional Type 1 Dendritic Cells in Expansion and Antitumor Efficacy of Adoptively Transferred Tumor-Specific T Cells

Using knockout and bone marrow chimera mouse models, Oba, Hoki, and Yamauchi et al. showed that CD40 and CD70, but not CD80/86, signaling on Batf3-dependent host cDC1s was essential for the expansion and antitumor efficacy of adoptively transferred gp100-specific CD8+ T cells (ACT) in mice bearing B16 tumors and treated with a vaccine (hgp100 peptide, TLR7 agonist, agonistic (ag) anti-CD40 Ab) and systemic IL-2. Administration of Flt3L prior to ACT with TLR3/agCD40-based vaccination induced and activated host cDC1s and enhanced the expansion of transferred T cells, which could be useful in a clinical setting.

Contributed by Lauren Hitchings

The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies

In a cohort of gastric and non-small cell lung cancer patients treated with anti-PD-1, responding patients had higher PD-1 expression on tumor-infiltrating CD8+ T cells and reduced PD-1 on effector Tregs. PD-1 expression on CD8+ T cells correlated with exposure to higher-affinity antigens. Anti-PD-1 mAb activated both PD-1+ Tregs and CD8+ T cells by restoring CD3/CD28 signaling and downstream ZAP70/AKT phosphorylation. The relative expression of PD-1 on CD8+ T cells and Tregs regulated proliferation in co-culture and tumor progression in vivo, and could predict anti-PD-1 patient responders better than prior biomarkers (AUC 0.933).

Contributed by Alex Najibi

Bispecific Targeting of PD-1 and PD-L1 Enhances T-cell Activation and Antitumor Immunity

Kotanides et al. developed a bispecific monoclonal antibody (LY3434172) that co-targets human (and cynomolgus nhp) PD-1 and PD-L1 with high specificity and comparable binding affinity to each parental antibody. LY3434172 induced bridging of PD-1- and PD-L1-expressing cells and enhanced T cell activation compared to each parental antibody and their combination. In xenograft tumor models (HCC827 and NCI-H292) complemented with human immune cells (T cells or PBMCs), LY3434172 demonstrated enhanced antitumor activity compared to each parental antibody and their combination at considerably lower doses.

Contributed by Shishir Pant

Immunomodulatory Activity of a Colony-Stimulating Factor-1 Receptor Inhibitor in Patients With Advanced Refractory Breast or Prostate Cancer: A Phase 1 Study

Autio et al. report the safety and clinical activity of an anti-CSF-1 receptor antibody (LY3022855) in patients with metastatic breast and castration-resistant prostate cancer (MBC and mCRPC). LY3022855 increased the levels of circulating CSF-1 and IL-34, and reduced circulating proinflammatory CD14DIMCD16BR monocytes. Out of 34 treated patients, 5 MBC and 3 mCRPC patients showed SD. Two MBC patients showed durable SD for over 9 months. Patients with SD had an increase in circulating LAG3+CD4+ and LAG3+CD8+ T cells and intratumoral immune activation-related gene expression. No unexpected treatment related adverse effects were seen.

Contributed by Shishir Pant

Lack of myeloid cell infiltration as an acquired resistance strategy to immunotherapy

Beyranvand Nejad et al. showed that suboptimal HPV16 E7 peptide vaccination of E7+ tumor-bearing mice induced specific T cell responses that mediated tumor regression, but allowed regrowth that could not be prevented by using anti-PD-1. Relapsed tumors expressed E7, were killed in vitro by T cells from vaccinated mice, and had changes in IL-6, TGFβ, TNFα, and p53 that were not causative of resistance. Vaccination of naive hosts bearing tumor cells from relapsed tumors did not induce regression despite CD8+ T cell infiltration. Attracting inflammatory myeloid effector cells, which non-responding lesions from HPV16-vaccinated patients lacked, restored vaccine response.

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.