Our major findings are that IRF2 expression was reduced in subsets of almost all human cancer types and that this reduction had functional consequences. When IRF2 expression was reduced, there was a corresponding reduction in the expression of downstream IRF2 target gene transcripts. This positive correlation was observed between IRF2 expression and that of almost all genes in the MHC I pathway, other IRF2-regulated genes, but not in several housekeeping genes that were examined. Remarkably these correlations were observed in almost all cancer types in the TCGA database. One of these cancer types, melanoma, was selected for further analysis. When IRF2 was knocked out of mouse or primary patient melanomas, there was also a reduction in the expression of IRF2 target genes, establishing a cause-and-effect relationship between IRF2 expression and these genes. In both human and mouse melanomas, loss of IRF2 led to resistance to CPI immunotherapy in preclinical models. Importantly, the IRF2 immune evasion phenotype could be reversed by treatment of cells with type I and II IFN. Moreover, the resistance of IRF2-deficient melanomas to immunotherapy could be restored by treatment with a type I IFN inducer in combination with CPI. In melanomas that were deficient in both IRF1 + IRF2, IFN treatment failed to restore the MHC I pathway and reverse the resistance to CPI, indicating that the beneficial effects of the IFN treatment were mediated through the substitutive activity of the transcription factor IRF1. These results elucidate a mechanism that underlies cancer immune evasion through loss of IRF2 expression, which is reversible with currently available biologics, and is likely applicable to many cancers.
The mouse and human melanomas we examined were both ones that were responsive to CPI, but these responses were partial, i.e., CPI treatment slowed their growth and survival was extended, but this did not result in tumor elimination. This is similar to what has been seen in previous studies with these same cancer cells in mice 11,20–23. We believe that these are appropriate preclinical models as such partial responses are what is observed in many CPI-treated cancer patients, including ones with melanomas 24–26. For tumors like B16 melanoma, this may reflect the fact that they are quite aggressive and likely developed some ability to evade immune responses. Against this baseline, loss of IRF2 clearly converted these cancers to being non-responders to CPI. Since our transcriptomic analyses showed that IRF2 levels were “rate limiting” for expression of MHC I pathway components and caspase 7 in most patient cancers, our treatment results predict that patients whose cancers have reduced levels of IRF2 transcripts, which we showed can be frequent, will similarly become more resistant to CPI. This prediction needs to be tested in clinical studies.
Loss of IRF2 may confer resistance to CPI is several ways that are not mutually exclusive. One way is through down regulation of MHC I pathway components. IRF2 positively regulates transcripts for almost all components in the MHC I pathway 6,10. Our transcriptomic analyses here suggest that this is the case in subsets of many human cancer types. Since as discussed above, IRF2 levels seem to be limiting, the reduction in IRF2 expression will affect the expression of multiple components of the pathway regulating MHC I expression with the net effect in the impairment of the MHC I antigen pathway expected to be additive. While we didn’t resolve which of the IRF2-regulated MHC I components became functionally limiting in these cancer cells, we had previously shown that in IRF2 null cells, TAP and ERAP1 functions (peptide transport into the ER and subsequent peptide trimming) are inhibited, as is overall antigen presentation 6. In any case, the important point is that the net effect of the reduction of MHC I pathway transcripts is a decrease in the number of peptide-MHC I complexes on the cell surface. This should make it harder for CD8 T cells to recognize the IRF2low cancers. Where examined, low MHC I levels in cancers have been associated with poorer responses to CPI 5.
Another way that loss of IRF2 might confer resistance to CPI is through a reduction in caspase 7. This is because as discussed above, reductions in caspase 7 could impair the ability of CD8 T cells to kill cancer cells with granulysin. However, this would not cause resistance in our mouse melanoma because murine CD8 T cells lack granulysin. Yet another way that IRF2 could lead to immune evasion is by decreasing the repression of PDL-1. However, while this can occur in some mouse and human cells 6,10, we did not observe this effect in the present study. Thus, IRF2 repression of PDL-1 may be cell line specific. Finally, it is possible that a reduction in other IRF2-dependent processes (e.g., gasdermin D), may contribute to the immune evasion phenotype and resistance to CPI.
NK cells can also kill tumor cells. But, they recognize cells that do not express receptors MHC I and recognition of surface MHC I molecules inhibits NK cytotoxicity. Consequently, some cells lacking MHC I molecules are killed by NK cells27. In a preliminary experiment we found that the growth rate of IRF2-sufficient versus IRF2-null B16 cells was identical in mice that were depleted of NK cells (Supp. Figure 5B). Perhaps this is because the IRF2-deficient tumor cells are able to evade NK cells because they still expresses some MHC I molecules and/or have decreased caspase 7, however further studies are needed to determine whether and to what extent NK cells can recognize and control IRF2-deficient cancers. Finally some additional as yet unknown targets of IRF2 in CD8 or NK cell may be involved.
IRF-2 is a transcription factor with many gene targets and thereby plays diverse roles. IRF-2 has been implicated in the regulation of cell growth and differentiation in various cell types influencing the expression of genes involved in cell cycle regulation, apoptosis, and differentiation processes 28. We couldn’t detect any proliferation and growth differences in IRF2-deficient mouse melanoma cell line either in vivo or in vitro. On the other hand, the IRF2-deficient human patient melanoma grew more aggressively in both severely immunodeficient NSG mice and human hematopoietic stem cell transplanted, immunocompetent NSG mice. These observations may be related to earlier studies showing oncogenic effects of IRF2 suppression 29, although overexpression of IRF2 has also been reported to cause oncogenic transformation in pancreatic cancers and leukemias 30–32. Perhaps how IRF2 expression affects tumor growth depends on the particular cancer. Downregulation of IRF2 in human melanomas might be creating a double whammy via downregulation of MHC I antigen presentation to escape immune detection and releasing the brakes on cell proliferation at the same time. It will be of interest in future studies to examine whether this is the case in other tumors and experimental settings. Whether the IRF2 effects on the human melanoma growth contribute to CPI resistance to in our system is not resolved by our data, although other rapidly growing tumors like B16 do respond to CPI.
IRF2 is constitutively expressed in cells and our data showed that this expression is important for maintaining the activity of the MHC I pathway and expression of MHC I molecules. IRF2’s close relative, IRF1, is induced by type I and II IFN-stimulation and binds competitively to the same DNA motifs as IRF2. IRF1 and IRF2 are both activators of the MHC I pathway genes. Importantly we found that type I and II IFN stimulation of IRF2-null melanomas restored their MHC I pathway and MHC I molecule expression and that this was in part dependent on IRF1. Similarly, systemic treatment with a type I IFN inducer, poly I:C, reversed resistance of IRF2 null cells to CPI in vivo and this salutatory effect was also dependent on IRF1 in the cancer cells. This demonstrates that IFN induction of IRF1 can reverse the immune evasion consequences of the loss of IRF2. When both IRF1 and IRF2 were absent, type I IFN treatment could not restore MHC I expression or reverse resistance to CPI, confirming the concept that type I IFN can induce IRF1 and restore immunogenicity of the tumor cells and host cytotoxic response to them. These findings have some likely translational implications. Treatment of melanoma patients with pegylated IFNa2b as an adjuvant therapy post-surgical resection was approved by the FDA 33. Our results suggest a potential mechanism that might contribute to this agent’s efficacy in treating melanoma. Our results further suggest that in patients with IRF2low melanomas or potentially other IRF2low cancers, adding IFN treatment to CPI therapy might improve efficacy. In fact, a recent study found that prior treatment with pegylated-IFN-alfa-2b increased the effectiveness of adjuvant pembrolizumab (anti-PD-1 treatment) in patients with surgically removable advanced melanoma, although IRF1 and IRF2 status was not evaluated 34. Since type II interferon can also induce IRF1, and this cytokine was FDA-approved for other indications, future studies should examine the effects of type II IFN on melanomas that are IRF2-deficient and its potential as an adjuvant therapy with CPI.
Our studies suggest low levels of IRF2 may be responsible for some of the poor immunogenicity of many tumors and that in preclinical studies, type I or II interferons can induce tumor expression of IRF2, enhancing MHC I expression and lead to better rejection, including in combination with CPI treatment. Given our findings, in future studies it will also be of interest to examine whether IRF2 expression levels in tumor biopsies might be a biomarker for subsequent responsiveness to CPI either by itself or with other markers. Similarly, it would be on interest to determine whether a cancer’s levels of IRF1 and IRF2 might be biomarkers for tumors that might benefit IFN treatment to improve the efficacy of CPI.
FIGURE LEGENDS
Figure 1. (A) Spearman correlations between transcripts of IRF2 and those of MHC I pathway genes (β2M, Erap1/2, HLA-A, HLA-B and HLA-C, PDIA3, PSMB9-10, PSME1, TAP1/2, TAPBP and TAPBPL) in tumor tissue from patients with the indicated cancer types (TCGA abbreviations). (B-D) Gene expression correlation between IRF2 vs ERAP1, PSME1, PSMB9, and Casp7 transcripts in primary and/or metastatic melanomas. (E) Spearman correlations between IRF2 transcripts and IRF2-regulated genes (Casp7, CD274 and GSDMD) and housekeeping genes (CALR, GAPDH, PSMB-7) in tumor tissue from patients with the indicated cancer types (TCGA abbreviations). (A-E) Data comes from the TCGA RNAseq database and was analyzed with TIMER 35
Figure 2. Loss of IRF2 reduces the expression of MHC I pathway components in a primary human melanoma. (A) Diagram of gene editing of a human patient melanoma (AV17) from passage in NSG mice. After editing, NSG mice were injected s.c. with WT and IRF2KO tumors, and once the tumors were palpable, they were harvested and analyzed for: (B) the expression of MHC I and PDL1 molecules on the tumors was analyzed by flow cytometer (C) mRNA expression of the MHC I pathway components was analyzed by qPCR. Each dot represents a biological replicate. Statistical analysis was calculated by GraphPad Prism, **P < 0.01, ***P < 0.001.
Figure 3. Loss of IRF2 in the mouse melanoma cell line B16F0 reduces the expression of MHC I pathway components but has no effect on B16F0 tumor growth kinetics in NSG or C57BL/6 mice. (A) Diagram of the experimental setup and (B) In vitro mRNA expression levels of MHC I pathway components in B16F0WT wt (n = 4) vs IRF2KO (n = 3) were analyzed by qPCR. (C) Diagram of experiments testing the in vivo growth of WT vs IRF2KO B16F0 cells in NSG mice (n = 10) and C57BL/6 mice (n = 10) (D&E) Tumors from C were collected on day 15 and MHC I expression was analyzed by flow cytometry (D) and mRNA expression of the MHC I pathway components was analyzed by qPCR (E). Each dot represents a biological replicate and the curves on F show mean + SD. Statistical analysis was calculated by GraphPad Prism, **P < 0.01, ***P < 0.001.
Figure 4. IRF2-deficient human and mouse melanomas are resistant to CPI therapy. (A) Diagram of experiments testing the WT (n = 14) vs IRF2KO (n = 12) B16F0 in vivo tumor growth in C57BL/6 mice after isotype control or αPD1 treatment. (B) Tumor growth was recorded until the end of the experiment. (C&D) Tumors were collected on day 17 and MHC I expression was analyzed by flow cytometry (C) and mRNA expression of MHC I pathway components were analyzed using qPCR method (D). (E) Another group of C57BL/6 mice (n = 58) were subcutaneously injected with either WT (n = 29) or IRF2KO (n = 29) B16F0 cells and tumor growth was recorded for survival analysis. (F) Diagram of experiments testing the WT (n = 14) vs IRF2KO (n = 15) the A17 patient-derived human melanoma growth in NSG (n = 6) and NSG with HuHSC (n = 23) mice after isotype control or αPD1 treatment. (G) Tumor growth was recorded until the end of the experiment and (C) MHC I expression was analyzed by flow cytometry on day 55 and day 112. (C,D&H) Each dot represents a biological replicate and the curves on B and G show mean + SD. Statistical analysis was calculated by GraphPad Prism, **P < 0.05, **P < 0.01, ***P < 0.001.
Figure 5. Effect of IFNα on WT and IRF2 KO B16 melanomas. (A) MHC I levels on IRF2KO (n = 3) and wt cells (n = 3) after 24 hour stimulation with or without IFNα (histograms) and after withdrawal of IFN (line graph). (B) mRNA expression of MHC I pathway components in WT (n = 2) and IRF2KO cells (n = 2) after the IFN treatments. (C) Diagram of the experiment testing the effects of IFNα + poly(I:C) treatment ± anti-PD1 on wt vs IRF2KO B16 melanoma. (D&E) Day 15 post tumor injection, tumors were harvested and (D) mRNA expression of the MHC I pathway components was analyzed by qPCR and (E) the expression of MHC I molecules on the tumors was analyzed by flow cytometer. Each dot represents a biological replicate and the curves on A. and C. show mean + SD. (F) Another group of C57BL/6 mice (n = 40) were injected with WT (n = 20) and IRF2KO (n = 20) tumors for survival and tumor growth was recorded for survival analysis. Statistical analysis was calculated by GraphPad Prism, **P < 0.01, ***P < 0.001.
Figure 6. Transcription factor IRF1 substitutes for the loss of IRF2. (A) Same experimental design as 5C and 5D except, IRF1 mRNA expression was analyzed by qPCR. (B-E) EV, IRF1KO, IRF2KO and DKO (IRF1 + IRF2KO) B16F0 cells were stimulated with IFNα in vitro and analyzed by: (B) Western blot for IRF1 or ß-actin; (C). qPCR expression for MHC I pathway components (n = 2/group). (D&E) Surface MHC I levels after 0 or 100 ng/ml IFNα treatment for 24 h (n = 2/group).
Figure 7. B16 melanoma IRF1 + IRF2 double KO cells show impaired responses to IFN inducer Poly(I:C) plus CPI. (A) NSG mice (n = 10) and C57BL/6 mice (n = 12) were subcutaneously injected with WT, IRF2KO and IRF1 + IRF1KO cells and data display tumor growth in individual mice. (B) Tumors were collected on day 15 and the surface MHC I expression of C57BL/6 tumors was analyzed by flow cytometry. (C) C57BL/6 mice (n = 45), that were treated with poly(I:C) and aPD1 were injected with IFNα treated (10ng/mL, 24h) control WT vs IRF2KO IRF2 + IRF1 (double) KO B16 cells and tumor growth was followed. (D&E) On day 17 post tumor injection, n = 5 mice/group were sacrificed for: (D) mRNA expression analysis of MHC I pathway components using qPCR method and (E) the tumor cell surface expression analysis of MHC I molecules by flow cytometer. Each dot represents the average measurement of the individual tumors collected from mice. (F) Survival analysis of WT or IRF2KO or IRF1 + IRF2 KO tumors in C57BL/6 mice. This experiment was repeated twice. Tumor growth curve shows mean + SD (C).
Supp. Figure 1. Differential expression of IRF2 gene in tumor and corresponding normal tissue from patients with the indicated cancer types (TCGA abbreviations).
Supp. Figure 2. (A) A17 patient-derived human melanoma. was implanted into NSG (n = 9) and NSG with HuHSC (n = 15) mice. On day 35 (IRF2KO) and day 77 (EV) tumors were harvested and MHC I expression was analyzed by flow cytometry. (B-E) B16F0 EV or IRF2 KO cells were implanted into humanized C57Bl/6 mice and treated as indicated. Cell surface MHC I (C) and PDL1 (D) expression levels and tumor weight (E) of B16F0 EV vs IRF2KO tumors were quantified on day 15–17 in vivo. (F) shows corresponding tumor growth data.
Supp. Figure 3. (A) Effect of IFNα and IFNγ on WT and IRF2KO B16 melanomas. (B) MHC I levels on IRF2KO (n = 3) and WT cells (n = 3) after 24 hour stimulation with 10 ng/ml IFNα or IFNγ. (C) mRNA expression of MHC I pathway components in WT (n = 2) and IRF2KO cells (n = 2) after the IFN treatments.
Supp. Figure 4. (A) Corresponding histograms of Fig. 2B showing MHC I and PDL1 levels on IRF2KO (n = 5) and WT cells (n = 3) tumors in NSG mice. (B) Corresponding histograms of Fig. 3D showing MHC I levels on IRF2KO (n = 5) and WT cells (n = 5) tumors in NSG mice. (C) Corresponding histograms of Fig. 4C showing PDL-1 levels on IRF2KO (n = 5) and WT cells (n = 7) tumors in C57Bl/6 mice after aPD1 or isotype treatment.
Supp. Figure 5. (A) IRF1 and IRF2 mRNA expression levels in wt B16 melanoma cells with (n = 4) or without (n = 3) 10 ng/ml IFNα treatment for 24h in vitro. (B) In vivo growth of wt (n = 3) vs IRF2KO (n = 3) B16F0 cells in C57BL/6 mice that were treated with anti-mouse NK1.1 antibody (clone: PK136, BioXcell) every 3 days starting at day − 2.