1. Introduction
Type I and type II IFNs are cytokines primarily produced by virus-infected cells to initiate innate immune responses. Although they share many biological functions, they have distinct receptors. Type I IFNs signal through IFNAR1/2 heterodimeric receptors, activate Tyk2 and JAK1, and phosphorylate STAT1/2, which form complexes with IRF9, translocate to the nucleus, and activate expression of interferon-stimulated genes (ISG). Type II IFN (IFN-γ) also has a heterodimeric receptor, IFNGR1/2, which, upon ligand binding, activates JAK1/2 and phosphorylates STAT1, resulting in nuclear translocation and activation of the ISGs. However, IFNRG1/2 can activate alternative signaling pathways as well (STAT4, ERK1/2, Pyk2, or CRK1) [
1].
There are over 2000 IFN-regulated genes (IRGs) known today, including growth factors (like VEGF, FGF, and ECGF), chemokines (such as MIB, EBI1, and IL-8), adhesion molecules (i.e., ICAM1, CD47, and ALCAM), MHC class I and II, apoptosis regulators (such as FAS and CASP4/8), signaling molecules (like IFI16 and STAT1/2), and several transcription factors (including IRF1-7, ISGF3G, MPB1, PBX3, and, interestingly, HIF1α). A comprehensive database of interferon-regulated genes is available on the Interferome website [
2].
Cytokine therapy of melanoma patients has a long history. As malignant melanoma is relatively resistant to chemotherapy and radiotherapy, for a long time, cytokine therapy was the option in adjuvant settings using type I IFN monotherapy or in combination with type II IFN or IL-2 [
3]. However, IFN therapy has very low efficacy: ~6% for 5-year disease-free survival and 3% for 5-year overall survival according to major recent meta-analyses [
4]. This low efficacy is improved in the case of high-risk ulcerated primary tumors, but disappears in the case of non-ulcerated ones [
3,
4]. Accordingly, malignant skin melanoma can be considered “by default” to be IFN resistant, although the contribution of the tumor or the host to this feature is not known. Accordingly, the type I IFN sensitivity/resistance issue in the case of melanoma was open for a long time.
Pioneer studies on unselected human melanoma cell lines identified the RCC1, IFI16, HOX2, and H19 signature of sensitivity and SHB and PKCζ as markers of resistance to type I IFN [
5]. Another study on human melanoma xenografts identified a five-IRG signature of sensitivity comprising MxB, leu-13, Kip1/p27, Rig-E, and BST-2 [
6]. Type I IFN signaling is involved in the regulation of oncogene-induced senescence [
7], therefore it was studied in BRAF-mutant melanocytes and melanomas in animal models. The role of type I IFNs in the carcinogenesis and the progression of BRAF-mutant melanoma was analyzed in a genetically manipulated mouse model where IFNAR1 was knocked out [
7]. Data indicated that melanoma carcinogenesis was promoted in the IFNAR1 deficient host and the resulting tumors were spontaneously metastatic. Restoration of IFNAR1 signaling in BRAF mutant melanoma cells attenuated tumor growth in vivo. However, analysis of IFN-treated human melanoma tumor samples showed only a trend of lower IFNAR1 with a poorer outcome of patients, suggesting that the host may also participate in the regulation of IFN signaling [
7,
8].
Immune checkpoint inhibitor (ICI) therapy fundamentally changed the management of melanoma patients, significantly improving the five-year survival [
9]. However, there is no useful predictive marker of efficacy and data indicate that this treatment does not work in ~50% of patients [
9,
10]. Predictive markers of ICI therapy in solid tumors have been developed and are widely used today [
11]. Unfortunately, in melanoma, the predictive role of the PD-L1 expression is controversial [
12], microsatellite instability is very rare [
13], and only a high tumor mutational burden is clinically relevant [
14]. Clinical research revealed that type II IFN signaling is necessary for efficacy of anti-CTLA-4 and anti-PD-1 immunotherapy [
15]. Another analysis revealed that an established IFN-γ gene expression signature score, together with the tumor mutation burden, is a powerful predictor of the efficacy of immunotherapy of melanoma patients [
16]. Using experimental melanoma models and anti-PD-1 treated patient-derived tumor tissues, it was revealed that maintained type I IFN signaling is also a necessary element of the efficacy of immunotherapy [
8]. Analysis of the gene expression of melanoma tissues during CTLA-4 and high dose IFN-α2b combination therapy revealed a pro-inflammatory gene signature as a predictor of response and efficacy [
17]. These data all point to the importance of type I IFN signaling in ICI therapy response, but do not differentiate between the stromal and tumor components. In the case of type II IFN signaling, tumor intrinsic responses have been observed, mainly consisting of WNT and MYC signaling component, as well as the components of the antigen presentation machinery [
15]. A study performed on a large melanoma database treated with anti-CTLA-4 therapy revealed that homozygous deletion of type I IFN genes is significantly associated with resistance [
18], further supporting the notion that type I IFN signaling of tumor cells may play a significant role in melanoma progression.
3. Discussion
Here, we report an experimental model where a novel IFN-α2-resistance gene expression signature (GES) was defined. IFN resistance was developed in vitro by long-term exposure to type I IFN of human melanoma cells HT168-M1. This IFN resistance was maintained in vivo when tumor cells were inoculated into SCID mice. The development of this resistance mechanism was independent of immune mechanisms, because it was developed during in vitro culturing and was maintained in immune suppressed rodent hosts; accordingly, it was intrinsic of melanoma cells. Using microarray analysis of the in vitro cultured melanoma cells, we defined 79 differentially expressed genes. Of these 79 genes, 24 belonged to IFN-regulated genes according to the Interferome analysis [
2]. Accordingly, the majority of the identified melanoma-related genes were not IFN-regulated. Clinical studies have indicated that upon recurrence after IFN-α therapy, melanomas overexpress STAT5 [
19], but this gene was not part of our list of differentially expressed genes. Similarly, none of the previously identified IFN resistance genes were present in this signature [
5,
6], most probably due to the non-immune mechanism of the development of resistance.
The PANTHER pathway analysis revealed that a significant component of the non-IRG part of DEGs belonged to the neuronal development pathways. Previous studies revealed that during melanoma progression, melanoma cells develop stem-cell like properties associated with the expression of SOX10, EZH2 transcription factors, EMT phenotypic switch regulators TWIST1/ZEB1, and the surface receptor CD172 [
20]. In the non-IRG gene list of IFN resistance, we found the
TYRP1 melanoma marker,
SSTR5 somatostatin receptor, and
RPE65 retinal pigment epithelial marker genes all overexpressed in resistant melanoma cells, suggesting that neural crest and melanocytic linage markers may have a role in developing IFN resistance. Furthermore, a recent analysis of melanomas exposed to anti-PD-1 therapy revealed alterations in the expression of melanocytic and neural crest-related genes [
21].
Our in silico analysis of IFN-treated melanoma tissues of TCGA revealed the differential expression of five members of our 79 IFN-res DEGs in IFN-treated melanoma tissues: IRG
SOX4 and non-IRGs
WFDC1,
BCAN,
RPE65, and
MAPT. The SOX4 transcription factor is reported to be upregulated in melanoma believed to be involved in metabolic rewiring [
22]. WFDC1 is a tumor suppressor frequently lost in breast and prostate cancers, hepatocellular carcinoma, and Wilms’ tumor. In a significant proportion of melanomas, WFDC1 is downregulated by hypermethylation and has been shown to inhibit expression of DKK1, a known WNT signaling inhibitor [
23].
DKK1 is part of the non-IRG DEGs, where it is significantly downregulated compared to
WFDC1, which is the most upregulated one. It is of note that in our experimental IFN resistance models,
WFDC1 was consistently found to be differentially expressed in IFN resistant and sensitive melanoma cells or tumors. BCAN (brevican) is a chondroitin sulphate proteoglycan of the ECM, with no data on its role in melanoma. RPE65 was shown to be expressed by nevi, but downregulated in melanoma [
24]. The upregulated HOXC11 is also a transcription factor, regulating the expression of linage marker S100b in melanoma [
25].
MAPT codes for the tau protein involved in Alzheimer and Parkinson’s diseases. Interestingly, recently, a connection between neurodegenerative diseases and melanoma was raised, demonstrating an accumulation of amyloid in melanoma metastases [
26].
Although the IFN-treated TCGA cohort of melanoma was very small, having clinical response data only for eight patients, we also tested the predictive power of components of our IFN-res DEGs. This analysis revealed four genes expressed significantly differently in responder patients compared to nonresponders: non-IRGs
WFDC1,
HSBP7,
HSF1, and the only IRG,
MT2A. It is of note that marker genes of IFN therapy and predictive genes of IFN therapy efficacy only overlapped by one gene,
WFDC1. This IFN therapy predictive gene set contained two members of the heat shock protein family,
HSPB7 and
HSF1. While there were no data on the role of HSPB7 in melanoma, HSF1 was shown to be upregulated in melanoma due to the loss of FBX7, and was shown to be involved in regulating the metastatic potential [
27]. The nearly significant TDP52L is a regulator of MAP3K5 protein kinase and has been shown to be involved in cell proliferation of melanoma cells [
28]. The IRG, MT2A is a metallothionein protein responsible for heavy metal ion detoxification. MT2A and other family members were shown to be overexpressed in melanoma and were associated with increased macrophage density of TME [
29].
Previous studies identified IFN signaling as the key predictive mechanism of the ICI therapies of melanoma. A tumor microenvironment-specific IFN-related ICI resistance signature was defined [
30,
31], as well as a tumor cell associated one [
32]. The tumor-specific component of the ICI resistance was due to genetic loss of
IFNGR1/2 and
JAK2, and amplification of IFN signaling inhibitors
SOCS1 and
PIAS4 [
32]. Other studies found downregulation or loss of heterozygosity of
HLA-B [
33],
B2M [
34],
JAK1 [
35], and
SERPINB3/4 mutations [
36] as markers of ICI resistance. However, a recent analysis defined a predictive 30-gene IFN-γ pathway expression signature of anti-CTLA-4 therapy, which contained three genes of the IRG component of our DEGs—
CAMK2D,
MT2A, and
HSP90AB1 [
37].
Analysis of the four most upregulated and four most downregulated genes of the in vitro obtained DEGs for their predictive power for response to ICI treatment identified four genes—
WFDC1,
EFNA3,
DDX10, and
PTBP1—with a significant predictive potential, and none of them were IRGs. We also analyzed the stability of these differentially expressed genes in vitro and in vivo using melanoma xenografts. We found that only a 13-gene subset of IFN resistance DEGs were stable in vitro, containing IRGs
SOX4,
UCP3,
DEK, and
HSPA1B. Furthermore, in two independent studies, we found that only a small subset of genes of the DEGs was present in vivo in xenografts consisting of a 4-gene core containing IRGs
IFI27 and
CDCA4 and two non-IRGs
AQP1 and
CDKL3. When the predictive power of all of these genes was tested on an ICI-treated melanoma patient cohort, 11 out of the 13 in vitro stable (containing IRGs
SOX4,
DEK, and
HSPA1B) and
AQP1 and
CDCA4 in vivo stable DEGs were differentially expressed in the tumors of ICI responder melanoma patients. In this way, we defined a 17-gene core the IFN resistance DEGs of melanoma, which all had a predictive potential for the ICI response of melanoma patients (
Table 7,
Table 8 and
Table 9).
EFNA3 is a hypoxia regulated gene and is a GPI-anchored ligand for the EPH receptors involved in cell adhesion and motility. It is a negative prognostic factor of gastric, ovarian, and lung cancers [
38,
39,
40]. DDX10 is an RNA helicase that is frequently lost in ovarian cancer [
41] and is a poor prognostic factor in osteosarcoma [
42]. PTBP1 is an RNA-binding protein involved in splicing. In dendritic cells, it was found that PTBP1 regulates the expression of several IFN-regulated genes [
43]. PTBP1 is expressed by melanoma stem cells [
44] and it has been shown to regulate CD44v6 expression in melanoma brain metastases [
45]. CDCA4 is an interferon-regulated E2F-type transcription factor involved in cell cycle regulation. In melanoma, miR-15a and miR-29c-3p are regulators of CDCA4, which is involved in controlling cell proliferation, invasion, and apoptosis [
46,
47]. AQP1 is a water channel protein, a hypoxia-regulated gene involved in various biological processes. AQP1 was found to be overexpressed in BRAF-mutant melanoma tumors and was shown to be a negative prognostic factor [
48].
It is of note that the in vitro stable IFN resistance 11-core DEGs contained two melanoma oncogenes,
SOX4 and
DEK (transcription factors); the former was reported to be downregulated, while the latter was found to be upregulated during melanoma progression [
49,
50]. Furthermore, this core-DEG also contained two neuronal genes,
NPTXR and
SSTR5, suggesting that melanoma stem cell properties might also have a role in ICI therapy resistance [
21]. Last, but not least, this core-DEG also contained two proteoglycans, BCAN (the chondroitin sulphate proteoglycan brevican) and SDC2 (the heparan sulphate proteoglycan, syndecan2). SCD2/syndecan2 was shown to be involved in the regulation of the migratory potential of melanoma cells [
51]. Furthermore, recently, proteoglycans were shown to be involved in the adaptive immune escape of experimental melanoma, where GUSB glucuronidase plays a role as a novel oncosuppressor [
52].
What could be the connection between type I IFN resistance and response to ICI therapy in melanoma? Type I IFN therapy was and is still is part of the management of melanoma patients [
3,
4]. From this perspective, it is of note that the IFN therapy and the ICI therapy predictive genes of our IFN resistance DEGs overlapped by three genes,
WFDC1,
SOX4, and
BCAN, strongly suggesting a connection between the two pheno-/geno-types of melanoma: IFN and ICI resistance. Progression of the disease after IFN therapy could be interpreted as development of IFN resistance in treated patients. It is tempting to speculate that melanomas that progressed after type I IFN therapy may respond differently to ICI therapies compared to those where such a therapy was not administered previously. Genetic analysis of the progressed melanomas previously treated with type I IFN compared to those who were not could reveal such a possible connection.