Next Article in Journal
The CCL5/CCR5 Axis in Cancer Progression
Next Article in Special Issue
High PD-L1/CD274 Expression of Monocytes and Blood Dendritic Cells Is a Risk Factor in Lung Cancer Patients Undergoing Treatment with PD1 Inhibitor Therapy
Previous Article in Journal
Cancer Immune Therapy for Philadelphia Chromosome-Negative Chronic Myeloproliferative Neoplasms
Previous Article in Special Issue
Purine Metabolites in Tumor-Derived Exosomes May Facilitate Immune Escape of Head and Neck Squamous Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy

by
Annelisa M. Cornel
1,†,
Iris L. Mimpen
1,† and
Stefan Nierkens
1,2,*
1
Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
2
Princess Máxima Center for Pediatric Oncology, Utrecht University, 3584 CS Utrecht, The Netherlands
*
Author to whom correspondence should be addressed.
These authors have contributed equally to this work.
Cancers 2020, 12(7), 1760; https://doi.org/10.3390/cancers12071760
Submission received: 11 June 2020 / Revised: 29 June 2020 / Accepted: 29 June 2020 / Published: 2 July 2020

Abstract

:
In recent years, major advances have been made in cancer immunotherapy. This has led to significant improvement in prognosis of cancer patients, especially in the hematological setting. Nonetheless, translation of these successes to solid tumors was found difficult. One major mechanism through which solid tumors can avoid anti-tumor immunity is the downregulation of major histocompatibility complex class I (MHC-I), which causes reduced recognition by- and cytotoxicity of CD8+ T-cells. Downregulation of MHC-I has been described in 40–90% of human tumors, often correlating with worse prognosis. Epigenetic and (post-)transcriptional dysregulations relevant in the stabilization of NFkB, IRFs, and NLRC5 are often responsible for MHC-I downregulation in cancer. The intrinsic reversible nature of these dysregulations provides an opportunity to restore MHC-I expression and facilitate adaptive anti-tumor immunity. In this review, we provide an overview of the mechanisms underlying reversible MHC-I downregulation and describe potential strategies to counteract this reduction in MHC-I antigen presentation in cancer.

1. Introduction

In recent years, major advances have been made in cancer immunotherapy, thereby drastically improving the prognosis of cancer patients. Several types of Food and Drug Administration (FDA)-approved immunotherapies, such as checkpoint inhibitors (CPI), chimeric antigen receptor (CAR) T-cells, and dendritic cell vaccines, aim to boost T-cell-mediated cytotoxicity to combat cancer. These treatments led to increased survival chances, particularly for patients suffering from hematological cancers, but translation to the solid tumor setting was found difficult. Additional immune escape mechanisms, including immune checkpoint expression, induction of immunosuppressive immune subsets (e.g., regulatory T-cells and myeloid-derived suppressor cells (MDSCs)), loss of immunogenic antigens, and decreased antigen presentation allow these tumors to evade anti-tumor immunity (reviewed by Sharma et al. [1]), posing serious challenges to overcome in order to improve therapy response.
One way in which tumors can avoid tumor-associated antigen presentation, and therewith T-cell-mediated cytotoxicity, is the downregulation of surface display of major histocompatibility complex (MHC) class I, a crucial factor in the initiation of an adaptive immune response. The importance of MHC-I downregulation in immune evasion is substantiated by the observed correlations between MHC-I expression on tumor cells and the amount of tumor infiltrating lymphocytes (TILs) in several cancers [2,3]. Furthermore, several groups have reported impaired MHC-I antigen processing and presentation as a predictor of (acquired) resistance to CPI therapy [4,5,6,7,8] and adoptive cell therapy [9,10,11].
Downregulation of MHC-I has been described in 40–90% of human tumors [9,12,13,14,15,16,17,18,19,20], often correlating with worse prognosis [18,21,22,23,24,25,26,27,28,29]. Both adult and pediatric tumors are able to reduce MHC-I surface display by the use of different regulatory mechanisms. Where adult tumors downregulate MHC-I expression in order to escape from the immune system, pediatric cancers, such as neuroblastoma, often arise from embryonic tissues that intrinsically lack immunological features, potentially explaining the low expression of MHC-I in these cancer types [30].
The intrinsic reversible nature of most MHC-I dysregulations provides an opportunity to restore MHC-I antigen presentation and facilitate adaptive anti-tumor immunity. This review aims to provide an overview of the mechanisms underlying reversible MHC-I downregulation and demonstrate potential therapeutic targets to induce MHC-I expression and improve T-cell-mediated cytotoxicity in cancer.

2. Dysregulation of MHC-I Expression in Cancer

T-cell receptors (TCRs) of CD8+ T-cells can only bind to their targets in the context of MHC-I, which is expressed on all nucleated cells. MHC-I presents endogenous antigens, a process important for reporting intracellular changes, for example caused by viral infections or malignant transformation, to the immune system in order to initiate a CD8+ T-cell response. The heterodimer MHC-I consists of a heavy chain, encoded by the human leukocyte antigen (HLA)-A, HLA-B, and HLA-C genes, and an invariant light chain called β2-microglobulin (β2M). The heterodimer requires stabilization by a peptide, which is loaded into the MHC-I peptide-binding groove by the antigen processing machinery (APM).
Antigen presentation in MHC-I context is a complex, multi-step process which can be dysregulated at many levels. MHC-I and β2M are synthesized in the endoplasmic reticulum (ER) and require stabilization by chaperone proteins (e.g., calreticulin, ERp57, and tapasin). Designated intracellular proteins are targeted for degradation by ubiquitination, after which they undergo proteasomal degradation into peptides. These peptides are subsequently translocated into the ER by the transporter associated with antigen processing (TAP) and bind to MHC-I directly or after further processing by ER aminopeptidases (ERAP1 and ERAP2). Interaction between tapasin and TAP allows translocation of peptides into the MHC-I binding groove, release of chaperone proteins, and stabilization of the MHC-I complex. Finally, the MHC-I-antigen complex travels via the Golgi apparatus to the cell surface, where antigen presentation to CD8+ T-cells can start. In cancer, one or several proteins in this complex pathway can be dysregulated, which may have major consequences on cell surface display of MHC-I (Figure 1).
Although downregulation of surface expression of MHC-I allows evasion from T-cell-mediated anti-tumor immunity, low MHC-I expression sensitizes cells to Natural Killer (NK) cell-mediated cytotoxicity. MHC-I functions as an inhibitory ligand for NK-cells by binding to inhibitory receptors, such as killer cell immunoglobulin-like receptors (KIRs), thereby dampening NK-cell activation. Accordingly, when MHC-I is downregulated, the inhibitory signals initiated by MHC-I are no longer present, leading to enhanced NK-cell activation and cytotoxicity [31]. Nonetheless, tumors have developed several mechanisms to escape NK-cell-mediated cytotoxicity. For example, tumors often produce factors, such as transforming growth factor β (TGF-β) and prostaglandin, that impair NK-cell function and block their infiltration into the tumor site [32]. Additionally, tumors may temporarily upregulate MHC-I expression in response to NK-cells, which allows them to avoid recognition by these cells [19,22,33]. As a result, tumors show plasticity in evading both NK- and T-cell-mediated cytotoxicity, thereby facilitating tumor immune escape. The importance of this plasticity is substantiated by the observation that colorectal cancer patients with reversible MHC-I downregulations have a worse prognosis compared to patients with irreversible MHC-I downregulations [22].
Dysregulated cell surface display of MHC-I complexes may be caused by genetic, epigenetic, transcriptional, and post-transcriptional alterations, which leads either to irreversible or reversible changes in MHC-I expression. Irreversible MHC-I defects are described in multiple types of cancer, including melanoma, head and neck squamous cell carcinoma (HNSCC), and lung, colorectal, bladder, laryngeal, and breast cancer [9,12,13,14,15,16,17] and arise due to structural genetic alterations, for example, in the class I heavy chain genes [12,13,14], β2M [9,15,16], and the TAP-encoding genes [17]. As MHC-I expression cannot be upregulated in tumors harboring this type of mutations, these irreversible defects will not be the main focus of this review. Reversible MHC-I dysregulations are characterized by the coordinated (post-)transcriptional downregulation of the HLA class I heavy chain, components of the APM, or β2M and have been observed in several types of tumors, including HNSCC, bladder cancer, and neuroblastoma [18,19,20]. The reversible nature of these pathway dysregulations makes them interesting targets in cancer immunotherapy.

3. MHC-I Expression Regulation

Three major transcription binding sites responsible for MHC-I heavy chain expression can be distinguished: an Enhancer A region, which can be recognized by NFkB; an interferon-stimulated response element (ISRE), which can be bound by interferon regulatory factor (IRF) 1; an SXY-module, which is recognized by NOD-like receptor family CARD domain containing 5 (NLRC5) [34]. Other APM genes are induced by the same set of transcription factors, resulting in the observation that the downregulation of APM players often coincides [35]. Accordingly, different pathways can lead to transcriptional activation of the MHC-I heavy chain-encoding genes as well as other genes responsible for the APM. In addition, these pathways can also act synergistically, magnifying the effect on MHC-I expression. By studying how MHC-I expression is normally regulated, we may be able to better understand the underlying mechanisms of MHC-I downregulation in tumors and find potential targets for cancer immunotherapy in order to reverse this downregulation.

4. Inducing MHC-I Expression in Cancer via NFkB Stabilization

The NFkB family consists of many inducible transcription factors, including NFkB1 (p50), NFkB2 (p52), RelA, RelB, and c-REL. During homeostasis, these proteins are sequestered in inactive cytosolic complexes by interaction with inhibitor of kB (IkB) family members (e.g., IkBα and p105). Upon pathway activation, IkB proteins are degraded, causing the release of NFkB transcription factors, allowing them to migrate to the nucleus to affect expression of target genes. Two major NFkB-inducing pathways can be distinguished: the canonical and the non-canonical NFkB pathway (Figure 2A,B) [36]. An elaborate description of pathway players important in this review can be found in Box 1. Although NFkB is constitutively active in most cancers [37], some tumors are able to downregulate NFkB signaling, which has been shown to impair MHC-I expression [35]. NFkB expression was found to be associated with favorable response to CPI therapy in patients with melanoma, indicating the potential of upregulating its expression to increase anti-tumor immunity [4,38]. To date, several regulators of the NFkB pathways have been described, revealing potential targets to enforce upregulation of MHC-I expression in cancer.
Box 1. NFkB-mediated upregulation of MHC-I.
The canonical NFkB pathway can be activated by stimulation of several immune receptors, including the TNFα Receptor (TNFR), Toll-like receptors (TLR), and cytokines receptors, by their ligands (e.g., TNFα, IL-1, or LPS). Upon receptor stimulation, TRAF2/6 and receptor-interacting protein 1 (RIP-1) are recruited, after which TRAF2/6 polyubiquitinates itself and RIP-1. In addition, TGF-β-activated kinase 1 (TAK1) is ubiquitinated and activated. TAK1 in turn is able to phosphorylate and activate the IkB kinase (IKK) complex (consisting of IKKα, IKKβ, and NF-kappa-B essential modulator (NEMO) (also known as IKKγ)). IKK needs to be recruited to the stimulated receptor before it can become phosphorylated, which is triggered by NEMO-mediated interaction of IKK with polyubiquitinated RIP-1 [39]. Subsequently, IKK phosphorylates IkB proteins, thereby targeting it for ubiquitination and degradation. Consequently, RelA-p50 and c-REL-p50 are released from their inactivating complexes, which results in translocation to the nucleus. Here they can activate transcription of their target genes, including MHC-I heavy chain- and other APM-encoding genes, by binding to kB enhancers in their promotors. A schematic overview of the canonical NFkB pathway can be found in Figure 2A [36].
Activation of the non-canonical NFkB pathway is initiated by the binding of a ligand (e.g., TNFα, CD40L, or B-cell activating factor (BAFF)) to one of the TNFR superfamily members. NFkB-inducing kinase (NIK) becomes activated, which in turn is able to phosphorylate and activate IKKα, one of the subunits of the IKK complex. IKKα subsequently phosphorylates the carboxy-terminal serine residues of p100, which is an IkB-like molecule that sequesters the NFkB transcription factor RelB in the cytosol, resulting in ubiquitination and degradation of the C-terminus of p100. As a result, p52 is generated, which forms a heterodimer with RelB, thereby allowing migration to the nucleus, resulting in induction of transcription of target genes. A schematic overview of the non-canonical NFkB pathway can be found in Figure 2B [36].

4.1. Positive Regulators of NFkB Expression

TNFα forcefully stimulates NFkB signaling and subsequent MHC-I expression. However, its use in cancer therapy is limited due to severe toxicities [40]. Alternatively, retinoids have been described to induce MHC-I upregulation in cancer, which is suggested to be the result of stimulation of NFkB signaling, even though the exact mechanism is still under debate. Studies in neuroblastoma and embryonic carcinoma cells revealed that retinoids increase MHC-I expression via increased expression of NFkB p50 and RelA [41,42]. In addition, Vertuani et al. [43] reported that upregulation of proteasome subunits (Latent Membrane Protein (LMP)-2, -7, and -10) and increased half-life of MHC-I complexes are responsible for increased MHC-I expression in neuroblastoma cell lines. Retinoids are currently being used to treat several types of cancer, including neuroblastoma and promyelocytic leukemia, and are known to induce differentiation, apoptosis, and inhibition of proliferation of tumor cells [44].
Other compounds reported to positively affect NFkB signaling of which to date the effect on MHC-I expression has not been assessed, are betulinic acid, and calcium/calcineurin combined with protein kinase C (PKC) antagonists. Betulinic acid upregulates canonical pathway activity via boosting the activity of IKK as well as phosphorylation and degradation of IkBα in multiple cancer cell lines, including neuroblastoma, glioblastoma, and melanoma [45]. In addition, it induces apoptosis, inhibits topoisomerase I activity, and suppresses angiogenesis in cancer [46,47,48,49,50]. Secondly, a study into latent HIV infections in CD4+ T-cells revealed that combining calcium/calcineurin and the protein kinase C (PKC) antagonist prostatin causes synergistic activation of NFkB through a similar mechanism as betulinic acid [51]. A third strategy would be to increase expression of the E3 ubiquitin ligase Nedd4 [52], which triggers polyubiquitination and proteasomal degradation of N4BP1 [53], a suppressor of NFkB. In addition, in B-cells, Nedd4 has been reported to induce ubiquitination and degradation of TNF receptor-associated factor (TRAF) 3 via the TNFR CD40, thereby inducing activation of both the canonical and non-canonical NFkB pathway [52]. However, to date, no Nedd4 stimulatory compounds have been described.
Nonetheless, dysregulation of factors affecting downstream signaling of NFkB-inducing pathways makes it questionable whether NFkB activity, and thus MHC-I expression, can be restored by upstream pathway activation. As a result, therapeutic intervention to inhibit negative regulators of NFkB may be a more effective strategy to explore.

4.2. Negative Regulators of NFkB Expression

The pediatric tumor neuroblastoma is well known for its low MHC-I expression. We have previously identified two major negative regulators of MHC-I via NFkB signaling in neuroblastoma: Nedd4 Binding Protein 1 (N4BP1) and TNFα-induced protein 3 interacting protein 1 (TNIP1) [54]. TNIP1 affects canonical NFkB activation, whereas N4BP1 exerts an effect on both canonical and non-canonical pathway activation.
N4BP1 interacts with several proteins involved in (de)ubiquitination, including NEDD4, Cezanne-1, A20, and Itch [53,54] and potentially also binds to polyubiquitin itself [55,56]. Polyubiquitin binding proteins, like N4BP1, can compete with NEMO for polyubiquitin binding, thereby directly antagonizing activation of canonical NFkB [56]. Besides this, N4BP1 interacts with the deubiquitinase (DUB) Cezanne-1, which functions in the deubiquitination and stabilization of TRAF3 [54] and subsequently modulates the activation of both the canonical [54] and non-canonical NFkB [57] by affecting the degradation of NIK and IkB, and the induction of c-REL ubiquitination and proteasome-mediated degradation [58].
TNIP1 (also known as A20-binding inhibitor of NF-kB (ABIN)-1) is known for its stimulatory effect on the NFkB inhibiting DUB A20 and inhibition of TNF-induced apoptosis and is upregulated in several types of cancer [59]. Similar to N4BP1, TNIP1 inhibits NFkB by impairing NEMO-mediated translocation of IKK to the receptor site. The exact mechanism remains unclear, but it is suggested to be via competing with NEMO binding to RIP-1 as well as via binding to a polyubiquitin group on NEMO itself [60]. TNIP1 is also able to prevent processing the IkB p105 into p50 [61] and to interact with the NFkB inhibiting DUB A20, stimulating deubiquitination of NEMO, thereby impairing IKK activation and thus canonical NFkB activation [62]. IL-17 treatment has been shown to induce proteasome-dependent downregulation of TNIP1 [63]. This resulted in NFkB activation, thereby suggesting its potential to upregulate MHC-I expression. However, IL-17 has been shown to play a key role in the promotion of tumor progression by inducing chronic inflammation, tumor cell proliferation, angiogenesis, and metastasis, which may majorly limit the use of this cytokine to induce MHC-I expression in cancer [64].
Hence, targeting N4BP1 or TNIP1 will result in strong activation of NFkB signaling, which in turn boosts MHC-I expression. Indeed microRNA (miR) 28-5p is an inhibitor of N4BP1 and has been demonstrated to act as a tumor suppressor in many cancer types, including colorectal cancer, renal cell carcinoma, and hepatocellular carcinoma [65,66,67]. In addition, MiR-1180 and miR-486 have also been demonstrated to induce NFkB activation via targeting of several inhibiting players of the NFkB pathway, including Cezanne, A20, and TNIP1-3 [68,69]. Nevertheless, it should be taken into account that these miRs have also been associated with cancer cell growth, survival, migration, and progression, emphasizing the need to further investigate the effect of these miRs in cancer [70,71].
The NFkB inhibiting DUB A20 is also involved in TNIP-independent regulation of NFkB signaling [72]. A20 can form a ubiquitin-editing complex together with the ubiquitin binding protein TAX1 Binding Protein 1 (TAX1BP1) and the E3 ubiquitin-protein ligase Itch. This complex can deubiquitinate RIP-1 and TRAF6, thereby inhibiting TAK1 activation as well as NEMO-mediated recruitment and activation of IKK. In addition, A20 can induce K48-polyubiquitination of RIP-1, thereby targeting it for proteasomal degradation [73]. TAX1BP1 is responsible for Itch recruitment to A20, whereas Itch controls the interaction between A20 and its substrates RIP-1 and TRAF6, enabling inactivation of these substrates. Knockout of either of these proteins results in inadequate NFkB inhibition, indicating that all three proteins are indispensable in the functioning of this ubiquitin-editing complex [74]. Itch has been shown to be upregulated in several types of cancer, such as breast cancer and neuroblastoma, in which it was shown to play a major role in cancer progression [75,76,77,78,79]. The antidepressant clomipramine, its structural homologue norclomipramine, and 1,4-naphthoquinone 10E have been shown to reduce tumor growth and enhance chemotherapy in multiple cancer cell lines, including breast, prostate, and bladder cancer lines, as well as in a multiple myeloma xenograft model [80,81]. In addition, clomipramine has been shown to induce MHC-I expression in a rat model of experimental allergic neuritis [82]. Controversially, N4BP1 has also been described to negatively regulate Itch by blocking the binding of Itch to its substrates [53]. However, as both N4BP1 and Itch play an important role in suppressing NFkB signaling, the potential for therapeutic inhibition of Itch remains elusive.
Another DUB enzyme, cylindromatosis (CYLD) plays a role in the inhibition of NFkB signaling through the removal of polyubiquitin motifs from NEMO and another important upstream protein in the NFkB signaling pathway called TRAF2 [83]. Several miRs have been reported to target CYLD, including miR-1288, -196, and -372-5p [84,85,86]. Recently, as reviewed by Farshi and colleagues, several DUB inhibitors have been developed for the treatment of cancer, all targeting different DUB enzymes that play distinct roles in the promotion of cancer [87]. To date, no specific DUB inhibitors have been described to specifically target Cezanne-1, A20, or CYLD. Nonetheless, aspecific DUB small molecule inhibitors, such as ubiquitin aldehyde (UbaI) have been described, which may suppress the activity of these NFkB targeting DUBs [87]. However, the use of aspecific inhibitors is limited due to the severe toxicity and simultaneous targeting of beneficial DUBs. Therefore, additional research should be conducted to develop specific DUB inhibitors. A patent (https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017109488, WO2017109488) describes cyanopyrrolidine derivates as specific inhibitors of Cezanne-1. In addition, as Cezanne-1 and A20 show sequence homology, it might potentially be possible to find cross-reactive DUB inhibitors to boost NFkB activity. However, in line with the above described miRs, Cezanne-1, A20, and CYLD are described in tumor suppressive as well as tumor progressive processes, which may be a counterindication for the use of inhibitors of these proteins in cancer [88,89].
Altogether, these studies show that there are multiple proteins that play a role in suppressing NFkB signaling, which may subsequently lead to hampered MHC-I expression. Several therapeutic interventions have been described, which could potentially trigger NFkB expression in cancer. A major contradiction we should be aware of is that the NFkB pathway is constitutionally active in a variety of tumors, playing an important role in many tumor-promoting processes, including inflammation, invasion, proliferation, angiogenesis, and metastasis [37]. This is further substantiated by the dual effect of most described proteins [53], miRs [68,69,70,71], and therapeutic interventions [53,64,75,76,77,78,88,89] affecting NFkB pathway induction on tumor-suppressing and tumor-promoting processes. It has been hypothesized that NF-kB inhibits tumor growth in cancers with a low mutational burden (early stages of cancer and potentially pediatric tumors), but that accumulation of mutations may lead to a loss of tumor suppressive function and the oncogenic features of NF-kB can become more dominant [90]. Therefore, we should carefully study the effects of NFkB pathway induction in NFkB-downregulated tumors as this could shift the balance from immune evasion towards tumor progression, thereby potentially even hampering the efficacy of cancer immunotherapy.

5. Inducing MHC-I Expression in Cancer via Restored IFN Signaling

In addition to NFkB, Interferons (IFNs) play a significant role in the induction of MHC-I expression. During homeostasis, the signal transducer and activator of transcription (STAT) proteins are present in the cytosol in their inactive form. Upon pathway activation, STATs become phosphorylated and dimerize, allowing them to migrate to the nucleus to affect expression of target genes. Both type I and type II interferon pathways are able to induce dimerization of STATs, thereby upregulating MHC-I expression using different signaling pathways (Figure 3A,B). An elaborate description of pathway players important in this review can be found in Box 2. IFNs play an important role in the regulation of antigen processing and presentation and are described to exert pro- and anti-tumorigenic effects in various types of cancer as extensively reviewed by Musella et al. and Castro et al. [91,92]. Downregulation of both type I and II IFN-mediated pathways have been described as mechanisms involved in resistance to CPI- and adoptive cell therapy in melanoma and lung cancer, indicating the potential of interference in these pathways to increase anti-tumor immunity [4,5,6,10,93,94,95,96]. Several factors affecting IFN pathway expression have been described, revealing potential targets to modulate MHC-I expression in cancer.
Box 2. Interferon-mediated upregulation of MHC-I.
The type II IFN pathway can be activated by stimulation of the IFNG receptor (consisting of IFNGR1 and IFNGR2) by IFNγ, allowing binding, phosphorylation, and activation of Janus Activated Kinase (JAK) 1 and JAK2. The intracellular domain of IFNGR1 is phosphorylated, creating a docking site for signal transducer and activator of transcription (STAT)1, which is phosphorylated by JAK1/2, forms homodimers, and translocates to the nucleus to activate target gene expression via binding to gamma-activated site (GAS) elements in the promotor regions of IFN-stimulated genes (ISG). One of the genes induced by STAT1 signaling is the transcription factor IRF1, which in turn is able to bind to the ISRE site present in the MHC-I promoter [34]. A schematic overview of the type II IFN pathway can be found in Figure 3A [97].
Activation of the type I IFN pathway is initiated by binding of type I IFNs (e.g., IFNα and IFNβ) to the IFNA receptor (consisting of IFNAR1 and IFNAR2), allowing binding, phosphorylation, and activation of JAK1 and tyrosine kinase 2 (TYK2). The cytoplasmic tail of IFNAR is phosphorylated, creating a docking site for STAT1-3, which are again phosphorylated by JAK1 to form homo and heterodimers. STAT1 homodimers can activate IRF1 expression as described for the type II IFN pathway. Additionally, STAT1/STAT2 forms a complex with IRF9 (called IFN-stimulated gene factor 3 (ISGF3), which is able to bind to the ISRE element in the MHC-I promotor. STAT3 homodimerization also occurs, however, this does not lead to MHC-I upregulation, but rather functions as a negative feedback loop to inhibit expression of pro-inflammatory genes. A schematic overview of the type I IFN pathway can be found in Figure 3B [98].

5.1. Positive Regulators of IFN Signalling

IFN signaling can be induced via treatment with IFN-inducing ligands, such as IFNα, IFNβ, and IFNγ. In addition, stimulation of several PRRs may result in downstream type I IFN production, thereby indirectly promoting MHC-I expression. IFNγ has been suggested to have the most potent effect on the expression levels of APM genes, including MHC-I, TAP, and ERAP, which may imply its beneficial effect in cancer immunotherapy [35,99,100]. However, in line with NFkB-inducing ligands, IFNs and PRR stimulation initiates a broad range of biological activities, thereby limiting its use in cancer therapy due to severe toxicities [101]. To avoid toxicities, targeted delivery of IFNγ to the tumor site may be of interest, for example by fusing it to an antibody specific for a tumor-associated antigen [102].
Another targeted approach would be a cellular NK-cell therapy strategy, as NK-cells are capable of either killing or upregulating MHC-I expression on MHC-I-lacking cells (via IFNγ secretion) [19,33,103]. The potential of NK-cell therapy is substantiated by observed correlations between the abundance of functional NK-cells and checkpoint inhibition efficacy in non-small cell lung cancer (NSCLC) [104,105]. However, several challenges remain, including anti-inflammatory responses within the tumor microenvironment (TME), which cause impaired NK-cell function and TME infiltration [19,33,106,107], as well as ex vivo expansion to generate sufficient NK-cell numbers, and in vivo persistence [108]. The suggested ability of tumors to both evade NK- and T-cell mediated cytotoxicity via plasticity in MHC-I expression is one of the anti-inflammatory mechanisms potentially decreasing efficacy of NK-cell therapy. Interestingly, several studies have observed a beneficial outcome for individuals with an inhibitory KIR genotype with lacking HLA-ligands (the ‘missing ligand’ genotype) undergoing cancer immunotherapy, as inhibitory KIRs without matching HLA-ligands cannot inhibit NK-cell cytotoxicity [109,110]. This led to the rationale of allogeneic, KIR genotype-mismatched NK-cell therapy strategies to optimize NK-cell cytotoxicity by decreasing HLA-dependent inhibition of NK-cells [111]. Besides this, it should be considered that the immune checkpoint programmed cell death-ligand 1 (PD-L1) is also shown to be upregulated by IFNγ [112,113]. As a result, IFNγ simultaneously induces MHC-I upregulation and T-cell suppression, thereby potentially creating a vicious circle of T-cell activation and inhibition.

5.2. Negative Regulators of IFN Signaling

The embryonic transcription factor double homeobox 4 (DUX4) has been shown to be upregulated in many cancer types, in which it causes downregulation of JAK1/2 and STAT1, thereby suppressing IFN target gene transcription, including MHC-I and other APM genes [114]. In line with this, DUX4 has been demonstrated to be significantly upregulated in non-responders to CPI, indicating that the DUX4-induced reduction in MHC-I expression results in decreased T-cell cytotoxicity in patients [114]. The exact mechanism in which DUX4 downregulates JAK1/2 and STAT1 remains to be elucidated. As DUX4 is studied in more detail in a disease called facioscapulohumeral dystrophy (FSHD), knowledge on treatment strategies could be gained from this disease. Bosnakovski et al. [115] reported that inhibition of p300, a histone acetyltransferase recruited by DUX4 to affect target gene expression, by a specific inhibitor, counteracted the effect DUX-4 overexpression in vitro and in vivo. Others reported before that p300 inhibition in multiple types of cancer led to suppressed proliferation [116]. However, whether p300 inhibition also restores IFN signaling remains to be elucidated. DUX4 downregulation was also observed when treated with p38 inhibitors in vitro and in vivo in FSHD models [117]. P38 inhibition has a favorable effect in cancer treatment, however, the wide variety of processes in which p38 is involved, including tumor suppressive processes, is a clear disadvantage of this inhibitor [118].
Lymphocyte adapter protein (LNK) has been reported to be able to negatively regulate IFN signaling via the induction of dephosphorylation of STAT1 [119,120]. LNK has been shown to be overexpressed in several solid tumors, including melanoma and ovarian cancer, and was found to be significantly increased in patients who did not respond to CPI therapy [120]. LNK has been reported to be targeted by miR-29b, miR-30-5p, miR-98, miR-181a-5p [121,122]. To date, no other therapeutic LNK inhibitors have been reported.
The peptidyl-prolyl isomerase Pin1 has been reported to induce ubiquitination and degradation of the type I IFN inducing transcription factor IRF3, which normally triggers a positive feedback loop upon type I IFN pathway stimulation [123]. Pin1 has been demonstrated to be elevated in multiple types of cancer, playing a role in stimulating several cancer-driving processes [124]. As Pin1 is involved in downregulating type I IFN signaling, its suppression may lead to enhanced MHC-I expression, thereby advancing T-cell-mediated cytotoxicity. Presently, several Pin1 inhibitors have been developed, such as miR-200b, miR-200c, and miR296-5p, the small molecules all-trans retinoic acid (ATRA) and KPT-6566, and the natural compound Juglone, which all showed anti-cancer activity in different types of cancer [125,126,127,128,129,130].
Several protein tyrosine phosphatases (PTPs) have been described to downregulate tyrosine phosphorylation in the JAK/STAT pathway, thereby interfering with induction of both type I and II IFN signaling. For example, PTPN1 dephosphorylates TYK2 and JAK2 [131], PTPN2 dephosphorylates JAK1, and PTPN11 (SHP2) has been demonstrated to inhibit phosphorylation of JAK1, STAT1, and STAT2 [132]. Tyrosine phosphatases have been shown to be elevated in multiple types of cancer, including breast, ovarian, gastric cancer, and glioma, in which they promote tumor growth, survival, and metastases [133]. To date, several efforts have been attempted in the development of tyrosine phosphatase inhibitors, including the PTPN11 inhibitor sodium stibogluconate, and the PTP1B inhibitor MSI-1436C, which are now being tested in clinical trials for various cancer types [133,134,135]. In addition, miR-155 has been reported to target several negative regulators of IFN signaling, including PTPN2, which has been shown to result in increased IFNγ production by T-cells within the TME, thereby promoting MHC-I expression [136]. In contrast, miRNA-155 has been demonstrated to be overexpressed in multiple types of cancer, playing a pivotal role in oncogenesis, which may limit its use in cancer [137].
The ubiquitin ligases RING finger protein 2 (RNF2) and Smad ubiquitination regulatory factor-1 (Smurf1) and the protein inhibitor of activated STAT (PIAS) all function via inhibiting transcription activation of STAT1 [138,139,140]. These proteins have been shown to be overexpressed in several types of cancer, including melanoma, gastrointestinal tumors, lymphoma, and pancreatic cancer, thereby playing an active role in cancer promotion [141,142,143,144,145,146]. Likewise, the ubiquitin ligase DCST1 is able to induce ubiquitination and degradation of STAT2, which results in hampered type I IFN signaling and has also been shown to be elevated in various cancers [147]. The small molecule inhibitors PRT4165 and A01 successfully inhibit, respectively, RNF2 and Smurf1 in vitro [148,149]. In addition, indirect inhibition of RNF2 was reported via inhibition of MEK-induced activation by the MEK-inhibitor trametinib [143]. To date, no DCST1 inhibitors have been described.
Finally, the protein kinase D2 (PKD2) has been shown to dampen type I IFN signaling via stimulating ubiquitination and endocytosis of IFNAR1, thereby causing rapid turnover of the type I IFN receptor [150]. PKD2 has been associated with multiple types of cancer, functioning by promoting tumor progression and blocking type I IFN signaling [151]. To date, several inhibitors of PKD2 have been developed, for example the small molecule inhibitor CRT0066101 and the pyrazolopyrimidine pan-PKD inhibitor SD-208, which both show anti-tumor activity in xenograft mouse models of various cancers [152,153].
Taken together, several proteins have been implicated in impaired IFN signaling in cancer. Accordingly, therapeutic intervention of these dysregulations may be beneficial when combined with immunotherapy to increase MHC-I expression and enhance T-cell-mediated cytotoxicity. However, many of these proteins have not been associated with MHC-I downregulation before, highlighting the need to investigate their effect on MHC-I expression. In addition, as stated for NFkB, IFNs are also reported to exert both pro- and anti-tumorigenic effects in various types of cancer [91,92], indicating the need to closely study these effects.

6. NLRC5-Mediated Upregulation of MHC-I

Only recently, NLRC5 was discovered as a third key regulator of MHC-I transcription [154]. NLRC5 is expressed in response to IFN-mediated signaling, in particular via IFNγ, by binding of a STAT1 homodimer to GAS in the NLRC5 promoter [154,155]. Interestingly, this STAT1 binding site in the NLRC5 promoter was shown to partially overlap with an NFkB binding site, suggesting that NLRC5 expression may be induced by NFkB as well [156]. Moreover, it has been suggested that the NLRC5 promoter contains an ISRE site, which may allow transcriptional activation by IRF1. Upon its expression, NLRC5 migrates to the nucleus, where it assembles with several proteins, including RFX, ATF1/CREB, and the NFY complex, resulting in the formation of the MHC enhanceosome. This complex is able to bind the SXY module in the MHC-I promoter, causing the induction of MHC-I expression. Additionally, NLRC5 is able to recruit transcriptional initiation and elongation factors, and histon-modifying enzymes, for example histon acetyltransferases and methyltransferases, which may further enhance transcriptional activation of MHC-I [82,157]. (Epigenetic) downregulation of NLRC5 expression has been observed as a mechanism of immune evasion in several types of cancer, including colorectal, ovarian, breast and uterine cancers [158,159]. NLRC5 has been reported to elicit anti-tumor immunity by enhancing antigen processing and presentation in melanoma [160]. In addition, NLRC5 expression correlates with response to CPI in melanoma (https://pubchem.ncbi.nlm.nih.gov/patent/US2017321285, US20170321285A1). Controversially, as also observed for NFkB- and IFN-mediated signaling pathways, NLRC5 has been reported to exert both pro- and anti-tumorigenic effects in various types of cancer (reviewed by Tang et al. [161]), indicating the need of careful evaluation of upregulation of NLRC5 in cancer. To date, no specific NLRC5 targeting compounds have been described. Nonetheless, as NLRC5 activation pathways largely overlap with the above described pathways, compounds inducing these pathways may also result in NLRC5 activation.

7. Inducing MHC-I Expression in Cancer via STAT3 Inhibition

As briefly touched upon in the section above, STAT3 becomes activated upon type I IFN signaling to function as a negative feedback loop to inhibit expression of pro-inflammatory genes, thereby contributing to balanced immune responses [98]. STAT3 is implicated in dampening a variety of immune responses, including inhibition of both NFkB- and IFN-mediated pathways. STAT3 is able to inhibit activation of the IKK-complex, thereby preventing phosphorylation and degradation of IkB, which results in sequestering of NFkB transcription factors in the cytosol.
STAT3 has been shown to play a key role in the promotion of cancer by mediating proliferation, survival, invasion, and metastasis [162]. STAT3 inhibition resulted in increased anti-tumor activity and superior responses to immunotherapy and immunogenic chemotherapy in several pre-clinical studies and trials [163,164,165,166,167,168,169,170,171,172]. Combinations of CPI and STAT3 inhibitors are currently tested in clinical trials [173]. STAT3 can be inhibited via upstream JAK inhibition as well as by direct inhibition of STAT3. Several specific STAT3 inhibitors have been described, including JSI-124 (cucurbitacin I) [163], static [166], indirubin [164], resveratrol [165], and the antibiotic nifuroxazide [170]. In addition, the multitarget tyrosine kinase inhibitors sorafenib and sunitinib are also reported to specifically target STAT3 phosphorylation and activation [167,168].
As STAT3 overexpression inhibits both NFkB- and IFN-signaling pathways, specific inhibition of STAT3 may be an interesting strategy to target downregulation of these pathways and potentially induce MHC-I upregulation. However, as is the case for other NFkB- and IFN-signaling pathway inhibitors, these pathway inhibitors also exhibit favorable effects, as for example indicated by the fact that STAT3 also elicits tumor-suppressive functions [174].

8. Inducing MHC-I Expression in Cancer via STING Induction

Stimulator of Interferon Genes (STING) is a DNA-sensing molecule which activates upon encountering foreign DNA by cyclic dinucleotide recognition [175]. Upon cyclic dinucleotide recognition, STING becomes activated and forms a complex with TANK-binding kinase 1 (TBK1), which in turn is able to phosphorylate and activate both RelA and IRF3, thereby stimulating both NFkB- and type I IFN-activation [176]. To date, several STING agonists have been developed to exploit this response in cancer. One example is the STING agonist SB 11285, which is currently tested in clinical trials in several solid tumors, including HNSCC and melanoma (ClinicalTrials.gov Identifier: NCT04096638), and previously showed strong anti-tumor immunity induction in mouse models [177].
As STING induced both NFkB- and IFN-pathway activation, agonizing its activity may be an interesting strategy to upregulate these pathways to potentially induce MHC-I upregulation. This is further substantiated by the correlation between STING expression and HLA-associated genes in neuroblastoma [178]. In addition, preclinical studies have reported superior effects of CPI therapy in combination with STING agonists [179,180]. Combining CPI with STING agonists is currently being tested in clinical trials [181]. The multi-facet involvement of STING in immune activation, however, underlines the need to study its effect to improve therapy outcomes while not compromising treatment safety.

9. Well-Known Oncogenic Pathways Affect MHC-I Expression

Various oncogenic pathways have been reported to affect expression of MHC-I, β2M, and other APM components in cancer, including the MAPK-, epidermal growth factor receptor (EGFR), HER2, c-MYC, and n-MYC pathway [182,183,184,185,186,187]. MAPK pathway activation is suggested to negatively influence MHC-I expression via decreased IRF1 activity and STAT1 expression [188]. The MEK inhibitors trametinib and cobimetinib have been demonstrated to enhance IRF1 expression and increased STAT1 phosphorylation in human keratinocytes [188]. Watanabe et al. [189] reported that treatment of a NSCLC cell line with trametinib also increased MHC-I expression in vitro. Another MEK inhibitor, selumetinib, increased MHC-I expression in papillary thyroid cancer cell lines [2]. In addition, upstream inhibition of BRAF by vemurafenib and dabrafenib also induced MHC-I and β2M upregulation in multiple melanoma cell lines [190].
Overexpression of the EGFR family member HER2/neu was shown to be inversely correlated with MHC-I expression in different types of cancer, including breast cancer, esophageal squamous cell carcinoma, and melanoma [191,192]. Additionally, overexpression of EGFR was associated with less potent responses to cancer immunotherapy in NSCLC and neuroblastoma [193,194]. EGFR activates PTNP11, which was mentioned above as a STAT1 inactivator [195]. PTNP11 also inhibits RAS GTPase-activating protein, which normally functions in downregulating MAPK signaling, thereby also contributing to impaired IFN signaling via the MAPK pathway [196]. A third mechanism in which EGFR signaling impairs MHC-I expression is the promotion of STAT3 activation [197]. HER2 in turn also induces proteasomal degradation of the tumor suppressor Fhit, which normally upregulates expression of MHC-I, APM components, and β2M via an as-yet unclear mechanism [142]. Hence, targeting tyrosine kinase receptors, for example by the anti-EGFR antibodies nimotuzumab and cetuximab, and the EGFR inhibitors afatinib, erlotinib, and gefitinib, results in enhanced expression of MHC-I and APM components in different cancer cell lines as well as in cancer patients [185,198,199,200,201,202].
Finally, oncogenes c- and n-MYC have both been associated with MHC-I downregulation [186,187,203]. N-MYC has been shown to decrease MHC-I expression in rat neuroblastoma cell lines through the inhibition of the NFkB transcription factor p50 [203]. On the contrary, Forloni et al. [204] showed that that the downregulation of MHC-I in human neuroblastoma cells was not caused by n-MYC. In line with this, we did not identify n-MYC as a negative regulator of NFkB [54]. However, one limitation of our study was the use of an early phase CRISPR/Cas 9 library, which impairs the ability to fully confirm the results found in previous studies. Recently, Yang and colleagues showed that c-MYC expression induction by the Wnt/B-catenin pathway is responsible for MHC-I downregulation in glioma [187]. This again hints towards involvement of MYC-family members in MHC-I expression regulation in cancer.
Altogether, by targeting these oncogenic pathways, we potentially will not only be able to impair tumor growth and proliferation, but also exhibit anti-tumor responses via increasing the immunogenicity of the tumor.

10. Chemotherapy- and Radiation-Induced MHC-I Expression

Certain chemotherapeutic therapy regimens are known to induce anti-tumor immune responses without inducing classical immunogenic cell death (ICD). Hodge and colleagues showed that both docetaxel-induced ICD-sensitive and resistant tumor cells expressed increased levels of APM components, including TAP2, calnexin and calreticulin [205]. In addition, immunochemotherapy combining IFNα and 5-fluorouracil treatment resulted in increased MHC-I expression via STAT1/2 activation in murine pancreatic cancer models [206]. Besides this, several topoisomerase inhibitors (e.g., topotecan, irinotecan, and etoposide), microtubule stabilizers (e.g., paclitaxel and vinblastine), cisplatin, and ionizing radiation elevate MHC-I surface expression, which is thought to be induced via NFkB stabilization and IFNβ secretion [207,208,209]. These studies indicate that cytostatic drugs may, besides direct induction of cell death, also be involved in decreasing tumor immune escape via upregulation of antigen presentation, thereby making these drugs interesting candidates for combination therapy to improve immunotherapeutic strategies in cancer.

11. Epigenetic Silencing Affecting MHC-I Expression

Another common reversible defect in MHC-I antigen presentation is the occurrence of epigenetic modulation, which may impair the transcription of MHC-I, APM components, β2M, or MHC-I regulatory proteins. Histon deacetylation (HDAC) has been reported to reduce expression of MHC-I and key components of the APM, such as the proteasome subunits LMP-2 and LMP-7, and TAP in multiple types of cancer, including neuroblastoma, glioma, Merkel cell carcinoma, cervical cancer, and melanoma [187,210,211,212]. HDAC inhibitors, of which several already are FDA approved (e.g., Romidepsin, Vorinostat and Panobinostat), show increased expression of MHC-I and APM components both in vitro and in vivo [187,210,211,213]. Multiple other HDAC inhibitors are currently tested in clinical trials either alone or in combination with other drugs in various types of cancer, including melanoma, breast cancer, and lung cancer [214].
Epigenetic modulation by DNA hypermethylation has also been reported to cause reduced expression of MHC-I and related genes, which may be reversed by treatment with DNA methyltransferase inhibitors (DNMTi). The DNMTis guadecitabine, 5-azacytidine, and decitabine increased MHC-I expression in breast cancer, melanoma, and neuroblastoma cell lines [54,215,216,217]. Interestingly, combinatory treatment with TNFα, IFNγ, or knockout of known inhibitors of NFkB signaling (TNIP and N4BP1) further exaggerated the effect of these epigenetic modulators [54,215,217]. Additionally, increased MHC-I expression was observed in breast cancer patients treated with a combination of DNMT and HDAC inhibitors [216], indicating the potential of epigenetic modification to reintroduce MHC-I expression in tumors.

12. Discussion

MHC-I-mediated antigen presentation is crucial for CD8+ T-cell cytotoxicity and is one of the key factors in endogenous adaptive immune response development as well as T-cell mediated immunotherapy efficacy in cancer. The often intrinsically reversible nature of these dysregulations provides an opportunity to restore MHC-I expression and therewith adaptive anti-tumor immunity. In this review, we highlight that MHC-I-mediated antigen presentation is a complex, multi-faceted process, which can be dysregulated at many levels. As APM players are often induced by the same set of transcription factors, downregulation of APM players often coincides. To identify therapeutic targets to increase immunotherapy efficiency in cancer, a better understanding of the underlying mechanisms of MHC-I downregulation in tumors is required. A summary of regulators of MHC-I expression and potential therapeutic strategies described in this review can be found in Table 1 and Figure 4.
Dysregulation of factors affecting the downstream signaling of MHC-I-inducing pathways makes it questionable whether pathway activity can be restored by upstream pathway activation. As a result, therapeutic intervention to inhibit negative regulators of these pathways, possibly combined with stimulation of positive regulators of these pathways, may be the most promising strategy to explore.
Downregulation of MHC-I is an important factor contributing to the immune evasion of tumors. However, as underlined by the observed resistance of solid tumors to CAR-therapy as well as by IFNγ-mediated upregulation of PD-L1 [112,113], immune evasion of tumors is a multifaced process. As a result, the authors hypothesize that combination therapy targeting several tumor-immunomodulatory processes simultaneously will be necessary to avoid immune evasion of tumors. Several combinations of immunomodulatory drugs, including combining CPI with STAT3 inhibitors or STING agonists, show promising results in (pre-)clinical studies [171,172,173,179,180,181].
An important contradiction in enhancing NFkB-, IFN-, and NLRC5-mediated MHC-I expression is the dual role of these pathways in cancer [37,91,92,161]. They have been described in both tumor-promoting as well as tumor-suppressive mechanisms in several types of cancer. The authors hypothesize that, depending on the activation status of these pathways, its function will either be to promote or suppress tumor progression. Hence, it is important to study the effect of (pharmacological) enhancement of these pathways in MHC-I lacking tumors, as overactivation might shift the balance from immune evasion towards tumor progression. However, the observation that several NFkB/IFN upregulatory drugs do show beneficial effects in the treatment of several types of cancer highlights the potential of these drugs in improving immunotherapy outcomes in cancer.

13. Conclusions

In conclusion, MHC-I antigen presentation is a complex process regulated by multiple pathways that can be pharmacologically targeted on multiple levels to increase pathway activation and trigger MHC-I expression in cancer. Increasing antigen presentation in MHC-I-downregulated tumors is key to increase adaptive anti-tumor immunity and improve (immuno)therapy efficacy.

Author Contributions

I.L.M. and A.M.C. performed the literature search and wrote the manuscript with critical comments from S.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Villa Joep Foundation, Grant number IWOV-Actief.51381.180034.

Acknowledgments

The authors thank Yvonne van de Grint for designing the figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Angell, T.E.; Lechner, M.G.; Jang, J.K.; LoPresti, J.S.; Epstein, A.L. MHC class I loss is a frequent mechanism of immune escape in papillary thyroid cancer that is reversed by interferon and selumetinib treatment in Vitro. Clin. Cancer Res. 2014, 20, 6034–6044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mina, M.; Boldrini, R.; Citti, A.; Romania, P.; D’Alicandro, V.; De Ioris, M.; Castellano, A.; Furlanello, C.; Locatelli, F.; Fruci, D. Tumor-infiltrating T lymphocytes improve clinical outcome of therapy-resistant neuroblastoma. Oncoimmunology 2015, 4, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shi, A.H.; Ho, L.-L.; Levine, S.; Yadav, V.; Cheah, J.; Soule, C.; Frederick, D.T.; Liu, D.; Boland, G.; Kellis, M. Epigenomic correlates of checkpoint blockade immunotherapy resistance. Proc. Am. Assoc. Cancer Res. Annu. Meet. 2019, 79. [Google Scholar] [CrossRef]
  5. Gettinger, S.; Choi, J.; Hastings, K.; Truini, A.; Datar, I.; Sowell, R.; Wurtz, A.; Dong, W.; Cai, G.; Melnick, M.A.; et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 2017, 7, 1420–1435. [Google Scholar] [CrossRef] [Green Version]
  6. Sade-Feldman, M.; Jiao, Y.J.; Chen, J.H.; Rooney, M.S.; Barzily-Rokni, M.; Eliane, J.P.; Bjorgaard, S.L.; Hammond, M.R.; Vitzthum, H.; Blackmon, S.M.; et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 2017, 8. [Google Scholar] [CrossRef]
  7. Chowell, D.; Morris, L.G.T.; Grigg, C.M.; Weber, J.K.; Samstein, R.M.; Makarov, V.; Kuo, F.; Kendall, S.M.; Requena, D.; Riaz, N.; et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018, 359, 582–587. [Google Scholar] [CrossRef] [Green Version]
  8. Lee, J.H.; Shklovskaya, E.; Lim, S.Y.; Carlino, M.S.; Menzies, A.M.; Stewart, A.; Pedersen, B.; Irvine, M.; Alavi, S.; Yang, J.Y.H.; et al. Transcriptional downregulation of MHC class I and melanoma de- differentiation in resistance to PD-1 inhibition. Nat. Commun. 2020, 11, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Benitez, R.; Godelaine, D.; Lopez-Nevot, M.A.; Brasseur, F.; Jiménez, P.; Marchand, M.; Oliva, M.R.; Van Baren, N.; Cabrera, T.; Andry, G.; et al. Mutations of the β2-microglobulin gene result in a lack of HLA class I molecules on melanoma cells of two patients immunized with MAGE peptides. Tissue Antigens 1998, 52, 520–529. [Google Scholar] [CrossRef]
  10. Lauss, M.; Donia, M.; Harbst, K.; Andersen, R.; Mitra, S.; Rosengren, F.; Salim, M.; Vallon-Christersson, J.; Törngren, T.; Kvist, A.; et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat. Commun. 2017, 8, 1–11. [Google Scholar] [CrossRef] [Green Version]
  11. Khong, H.T.; Wang, Q.J.; Rosenberg, S.A. Identification of multiple antigens recognized by tumor-infiltrating lymphocytes from a single patient: Tumor escape by antigen loss and loss of MHC expression. J. Immunother. 2004, 27, 184–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Maleno, I.; López-Nevot, M.; Cabrera, T.; Salinero, J.; Garrido, F. Multiple mechanisms generate HLA class I altered phenotypes in laryngeal carcinomas: High frequency of HLA haplotype loss associated with loss of heterozygosity in chromosome region 6p21. Cancer Immunol. Immunother. 2002, 51, 389–396. [Google Scholar] [CrossRef]
  13. Maleno, I.; Cabrera, C.M.; Cabrera, T.; Paco, L.; López-Nevot, M.A.; Collado, A.; Ferrón, A.; Garrido, F. Distribution of HLA class I altered phenotypes in colorectal carcinomas: High frequency of HLA haplotype loss associated with loss of heterozygosity in chromosome region 6p21. Immunogenetics 2004, 56, 244–253. [Google Scholar] [CrossRef] [PubMed]
  14. Maleno, I.; Romero, J.M.; Cabrera, T.; Paco, L.; Aptsiauri, N.; Cozar, J.M.; Tallada, M.; López-Nevot, M.A.; Garrido, F. LOH at 6p21.3 region and HLA class altered phenotypes in bladder carcinomas. Immunogenetics 2006, 58, 503–510. [Google Scholar] [CrossRef]
  15. Feenstra, M.; Veltkamp, M.; Van Kuik, J.; Wiertsema, S.; Slootweg, P.; Van den Tweel, J.; De Weger, R.; Tilanus, M. HLA class 1 expression and chromosomal deletions at 6p and 15q in head and neck squamous cell carcinomas. Tissue Antigens 1999, 54, 235–245. [Google Scholar] [CrossRef] [PubMed]
  16. Garrido, M.A.; Rodriguez, T.; Zinchenko, S.; Maleno, I.; Ruiz-Cabello, F.; Concha, Á.; Olea, N.; Garrido, F.; Aptsiauri, N. HLA class I alterations in breast carcinoma are associated with a high frequency of the loss of heterozygosity at chromosomes 6 and 15. Immunogenetics 2018, 70, 647–659. [Google Scholar] [CrossRef]
  17. Seliger, B.; Ritz, U.; Bock, M.; Huber, C.; Abele, R.; Tampé, R.; Sutter, G.; Sutter, G.; Drexler, I.; Ferrone, S. Immune escape of melanoma: First evidence of structural alterations in two distinct components of the MHC class I antigen processing pathway. Cancer Res. 2001, 61, 8647–8650. [Google Scholar]
  18. Meissner, M.; Reichert, T.E.; Kunkel, M.; Gooding, W.; Whiteside, T.L.; Ferrone, S.; Seliger, B. Defects in the human leukocyte antigen class I antigen-processing machinery in head and neck squamous cell carcinoma: Association with clinical outcome. Clin. Cancer Res. 2005, 11, 2552–2560. [Google Scholar] [CrossRef] [Green Version]
  19. Spel, L.; Boelens, J.J.; Van Der Steen, D.M.; Blokland, N.J.G.; van Noesel, M.M.; Molenaar, J.J.; Heemskerk, M.H.M.; Boes, M.; Nierkens, S. Natural killer cells facilitate PRAME-specific T-cell reactivity against neuroblastoma. Oncotarget 2015, 6, 35770–35781. [Google Scholar] [CrossRef] [Green Version]
  20. Romero, J.M.; Jiménez, P.; Cabrera, T.; Cózar, J.M.; Pedrinaci, S.; Tallada, M.; Garrido, F.; Ruiz-Cabello, F. Coordinated downregulation of the antigen presentation machinery and HLA class I/β2-microglobulin complex is responsible for HLA-ABC loss in bladder cancer. Int. J. Cancer 2005, 113, 605–610. [Google Scholar] [CrossRef]
  21. Squire, R.; Fowler, C.L.; Brooks, S.P.; Rich, G.A.; Cooney, D.R. The relationship of class I MHC antigen expression to stage IV-S disease and survival in neuroblastoma. J. Pediatr. Surg. 1990, 25, 381–386. [Google Scholar] [CrossRef]
  22. Watson, N.F.S.; Ramage, J.M.; Madjd, Z.; Spendlove, I.; Ellis, I.O.; Scholefield, J.H.; Durrant, L.G. Immunosurveillance is active in colorectal cancer as downregulation but not complete loss of MHC class I expression correlates with a poor prognosis. Int. J. Cancer 2006, 118, 6–10. [Google Scholar] [CrossRef] [PubMed]
  23. Turcotte, S.; Katz, S.C.; Shia, J.; Jarnagin, W.R.; Kingham, T.P.; Allen, P.J.; Fong, Y.; D’Angelica, M.I.; DeMatteo, R.P. Tumor MHC class I expression improves the prognostic value of T-cell density in resected colorectal liver metastases. Cancer Immunol. Res. 2014, 2, 530–537. [Google Scholar] [CrossRef] [Green Version]
  24. Andersson, E.; Villabona, L.; Bergfeldt, K.; Carlson, J.W.; Ferrone, S.; Kiessling, R.; Seliger, B.; Masucci, G.V. Correlation of HLA-A02* genotype and HLA class I antigen down-regulation with the prognosis of epithelial ovarian cancer. Cancer Immunol. Immunother. 2012, 61, 1243–1253. [Google Scholar] [CrossRef] [PubMed]
  25. Roemer, M.G.M.; Advani, R.H.; Redd, R.A.; Pinkus, G.S.; Natkunam, Y.; Ligon, A.H.; Connelly, C.F.; Pak, C.J.; Carey, C.D.; Daadi, S.E.; et al. Classical Hodgkin lymphoma with reduced β2M/MHC class i expression is associated with inferior outcome independent of 9p24.1 status. Cancer Immunol. Res. 2016, 4, 910–916. [Google Scholar] [CrossRef] [Green Version]
  26. Van Houdt, I.S.; Sluijter, B.J.R.; Moesbergen, L.M.; Vos, W.M.; De Gruijl, T.D.; Molenkamp, B.G.; Van Den Eertwegh, A.J.M.; Hooijberg, E.; Van Leeuwen, P.A.M.; Meijer, C.J.L.M.; et al. Favorable outcome in clinically stage II melanoma patients is associated with the presence of activated tumor infiltrating T-lymphocytes and preserved MHC class I antigen expression. Int. J. Cancer 2008, 123, 609–615. [Google Scholar] [CrossRef]
  27. Simpson, J.A.D.; Al-Attar, A.; Watson, N.F.S.; Scholefield, J.H.; Ilyas, M.; Durrant, L.G. Intratumoral T cell infiltration, MHC class I and STAT1 as biomarkers of good prognosis in colorectal cancer. Gut 2010, 59, 926–933. [Google Scholar] [CrossRef] [PubMed]
  28. Inoue, M.; Mimura, K.; Izawa, S.; Shiraishi, K.; Inoue, A.; Shiba, S.; Watanabe, M.; Maruyama, T.; Kawaguchi, Y.; Inoue, S.; et al. Expression of mhc class i on breast cancer cells correlates inversely with her2 expression. Oncoimmunology 2012, 1, 1104–1110. [Google Scholar] [CrossRef] [Green Version]
  29. Hanagiri, T.; Shigematsu, Y.; Kuroda, K.; Baba, T.; Shiota, H.; Ichiki, Y.; Nagata, Y.; Yasuda, M.; Uramoto, H.; So, T.; et al. Prognostic implications of human leukocyte antigen class i expression in patients who underwent surgical resection for non-small-cell lung cancer. J. Surg. Res. 2013, 181, e57–e63. [Google Scholar] [CrossRef]
  30. Spel, L.; Schiepers, A.; Boes, M. NFκB and MHC-1 interplay in neuroblastoma and immunotherapy. Trends Cancer 2018, 4, 715–717. [Google Scholar] [CrossRef]
  31. Anfossi, N.; André, P.; Guia, S.; Falk, C.S.; Roetynck, S.; Stewart, C.A.; Breso, V.; Frassati, C.; Reviron, D.; Middleton, D.; et al. Human NK Cell Education by Inhibitory Receptors for MHC Class, I. Immunity 2006, 25, 331–342. [Google Scholar] [CrossRef] [PubMed]
  32. Lee, J.-C.; Lee, K.-M.; Kim, D.-W.; Heo, D.S. Elevated TGF-β1 Secretion and Down-Modulation of NKG2D Underlies Impaired NK Cytotoxicity in Cancer Patients. J. Immunol. 2004, 172, 7335–7340. [Google Scholar] [CrossRef] [PubMed]
  33. Jonges, L.E.; Giezeman-Smits, K.M.; Van Vlierberghe, R.L.E.; Ensink, N.G.; Hagenaars, M.; Joly, É.; Eggermont, A.M.M.; Van de Velde, C.J.H.; Fleuren, G.J.; Kuppen, P.J.K. NK cells modulate MHC class I expression on tumor cells and their susceptibility to lysis. Immunobiology 2000, 202, 326–338. [Google Scholar] [CrossRef]
  34. Jongsma, M.L.M.; Guarda, G.; Spaapen, R.M. The regulatory network behind MHC class I expression. Mol. Immunol. 2019, 113, 16–21. [Google Scholar] [CrossRef] [PubMed]
  35. Lorenzi, S.; Forloni, M.; Cifaldi, L.; Antonucci, C.; Citti, A.; Boldrini, R.; Pezzullo, M.; Castellano, A.; Russo, V.; van der Bruggen, P.; et al. IRF1 and NF-kB Restore MHC Class I-Restricted Tumor Antigen Processing and Presentation to Cytotoxic T Cells in Aggressive Neuroblastoma. PLoS ONE 2012, 7, 1–8. [Google Scholar] [CrossRef]
  36. Sun, S.C. The non-canonical NF-κB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef]
  37. Chaturvedi, M.M.; Sung, B.; Yadav, V.R.; Kannappan, R.; Aggarwal, B.B. NF-κB addiction and its role in cancer: One size does not fit all. Oncogene 2011, 30, 1615–1630. [Google Scholar] [CrossRef] [Green Version]
  38. Wells, K.; Hintzsche, J.; Amato, C.M.; Tobin, R.; Vorwald, V.; McCarter, M.; Shellman, Y.; Tan, A.C.; Robinson, W. Investigating the role of NF- κB signaling and immune checkpoint blockade therapy in melanoma. Clin. Res. 2019, 90, abstract 5002. [Google Scholar]
  39. Ea, C.K.; Deng, L.; Xia, Z.P.; Pineda, G.; Chen, Z.J. Activation of IKK by TNFα Requires Site-Specific Ubiquitination of RIP1 and Polyubiquitin Binding by NEMO. Mol. Cell 2006, 22, 245–257. [Google Scholar] [CrossRef]
  40. Cai, W.; Kerner, Z.J.; Sun, J. Targeted Cancer Therapy with Tumor Necrosis Factor-Alpha. Biochem. Insights 2008, 15–21. [Google Scholar] [CrossRef] [Green Version]
  41. Segars, J.H.; Nagata, T.; Bours, V.; Medin, J.A.; Franzoso, G.; Blanco, J.C.; Drew, P.D.; Becker, K.G.; An, J.; Tang, T. Retinoic acid induction of major histocompatibility complex class I genes in NTera-2 embryonal carcinoma cells involves induction of NF-kappa B (p50-p65) and retinoic acid receptor beta-retinoid X receptor beta heterodimers. Mol. Cell. Biol. 1993, 13, 6157–6169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Farina, A.R.; Masciulli, M.P.; Tacconelli, A.; Cappabianca, L.; De Santis, G.; Gulino, A.; Mackay, A.R. All-trans-retinoic acid induces nuclear factor κB activation and matrix metalloproteinase-9 expression and enhances basement membrane invasivity of differentiation-resistant human SK-N-BE 9N neuroblastoma cells. Cell Growth Differ. 2002, 13, 343–354. [Google Scholar] [PubMed]
  43. Vertuani, S.; De Geer, A.; Levitsky, V.; Kogner, P.; Kiessling, R.; Levitskaya, J. Retinoids Act as Multistep Modulators of the Major Histocompatibility Class I Presentation Pathway and Sensitize Neuroblastomas to Cytotoxic Lymphocytes. Cancer Res. 2003, 63, 8006–8013. [Google Scholar]
  44. Matthay, K.K.; Reynolds, C.P.; Seeger, R.C.; Shimada, H.; Adkins, E.S.; Haas-Kogan, D.; Gerbing, R.B.; London, W.B.; Villablanca, J.G. Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: A children’s oncology group study. J. Clin. Oncol. 2009, 27, 1007–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kasperczyk, H.; La Ferla-Brühl, K.; Westhoff, M.A.; Behrend, L.; Zwacka, R.M.; Debatin, K.M.; Fulda, S. Betulinic acid as new activator of NF-κB: Molecular mechanisms and implications for cancer therapy. Oncogene 2005, 24, 6945–6956. [Google Scholar] [CrossRef] [Green Version]
  46. Selzer, E.; Pimentel, E.; Wacheck, V.; Schlegel, W.; Pehamberger, H.; Jansen, B.; Kodym, R. Effects of betulinic acid alone and in combination with irradiation in human melanoma cells. J. Invest. Dermatol. 2000, 114, 935–940. [Google Scholar] [CrossRef] [Green Version]
  47. Wick, W.; Grimmel, C.; Wagenknecht, B.; Dichgans, J.; Weller, M. Betulinic acid-induced apoptosis in glioma cells: A sequential requirement for new protein synthesis, formation of reactive oxygen species, and caspase processing. J. Pharmacol. Exp. Ther. 1999, 289, 1306–1312. [Google Scholar]
  48. Fulda, S.; Friesen, C.; Los, M.; Scaffidi, C.; Mier, W.; Benedict, M.; Nuñez, G.; Krammer, P.H.; Peter, M.E.; Debatin, K.M. Betulinic acid triggers CD95 (APO-1/Fas)- and p53-independent apoptosis via activation of caspases in neuroectodermal tumors. Cancer Res. 1997, 57, 4956–4964. [Google Scholar] [PubMed]
  49. Chowdhury, A.R.; Suparna, M.; Mittra, B.; Sharma, S.; Mukhopadhyay, S.; Majumder, H.K. Betulinic acid, a potent inhibitor of eukaryotic topoisomerase I: Identification of the inhibitory step, the major functional group responsible and development of more potent derivatives. Med. Sci. Monit. 2002, 8, 254–261. [Google Scholar]
  50. Kwon, H.J.; Shim, J.S.; Kim, J.H.; Cho, H.Y.; Yum, Y.N.; Kim, S.H.; Yu, J. Betulinic acid inhibits growth factor-induced in vitro angiogenesis via the modulation of mitochondrial function in endothelial cells. Japanese J. Cancer Res. 2002, 93, 417–425. [Google Scholar] [CrossRef]
  51. Chan, J.K.; Bhattacharyya, D.; Lassen, K.G.; Ruelas, D.; Greene, W.C. Calcium/calcineurin synergizes with prostratin to promote NF-κB dependent activation of latent HIV. PLoS ONE 2013, 8, e77749. [Google Scholar] [CrossRef] [PubMed]
  52. Fang, D.F.; He, K.; Wang, N.; Sang, Z.H.; Qiu, X.; Xu, G.; Jian, Z.; Liang, B.; Li, T.; Li, H.Y.; et al. NEDD4 ubiquitinates TRAF3 to promote CD40-mediated AKT activation. Nat. Commun. 2014, 5, 1–11. [Google Scholar] [CrossRef] [PubMed]
  53. Oberst, A.; Malatesta, M.; Aqeilan, R.I.; Rossi, M.; Salomoni, P.; Murillas, R.; Sharma, P.; Kuehn, M.R.; Oren, M.; Croce, C.M.; et al. The Nedd4-binding partner 1 (N4BP1) protein is an inhibitor of the E3 ligase Itch. Proc. Natl. Acad. Sci. USA 2007, 104, 11280–11285. [Google Scholar] [CrossRef] [Green Version]
  54. Spel, L.; Nieuwenhuis, J.; Haarsma, R.; Stickel, E.; Bleijerveld, O.B.; Altelaar, M.; Boelens, J.J.; Brummelkamp, T.R.; Nierkens, S.; Boes, M. Nedd4-binding protein 1 and TNFAIP3-interacting protein 1 control MHC-1 display in neuroblastoma. Cancer Res. 2018, 78, 6621–6631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Fenner, B.J.; Scannell, M.; Prehn, J.H.M. Identification of polyubiquitin binding proteins involved in NF-κB signaling using protein arrays. Biochim. Biophys. Acta Proteins Proteomics 2009, 1794, 1010–1016. [Google Scholar] [CrossRef] [PubMed]
  56. Nepravishta, R.; Ferrentino, F.; Mandaliti, W.; Mattioni, A.; Weber, J.; Polo, S.; Castagnoli, L.; Cesareni, G.; Paci, M.; Santonico, E. CoCUN, a novel ubiquitin binding domain identified in N4BP1. Biomolecules 2019, 9, 284. [Google Scholar] [CrossRef] [Green Version]
  57. Hu, H.; Brittain, G.C.; Chang, J.H.; Puebla-Osorio, N.; Jin, J.; Zal, A.; Xiao, Y.; Cheng, X.; Chang, M.; Fu, Y.X.; et al. OTUD7B controls non-canonical NF-κB activation through deubiquitination of TRAF3. Nature 2013, 494, 371–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Yang, X.D.; Sun, S.C. Targeting signaling factors for degradation, an emerging mechanism for TRAF functions. Immunol. Rev. 2015, 266, 56–71. [Google Scholar] [CrossRef] [Green Version]
  59. G’Sell, R.T.; Gaffney, P.M.; Powell, D.W. A20-binding inhibitor of NF-κB activation 1 is a physiologic inhibitor of NF-κB: A molecular switch for inflammation and autoimmunity. Arthritis Rheumatol. 2015, 67, 2292–2302. [Google Scholar] [CrossRef] [Green Version]
  60. Wagner, S.; Carpentier, I.; Rogov, V.; Kreike, M.; Ikeda, F.; Löhr, F.; Wu, C.J.; Ashwell, J.D.; Dötsch, V.; Dikic, I.; et al. Ubiquitin binding mediates the NF-κB inhibitory potential of ABIN proteins. Oncogene 2008, 27, 3739–3745. [Google Scholar] [CrossRef] [Green Version]
  61. Cohen, S.; Ciechanover, A.; Kravtsova-Ivantsiv, Y.; Lapid, D.; Lahav-Baratz, S. ABIN-1 negatively regulates NF-κB by inhibiting processing of the p105 precursor. Biochem. Biophys. Res. Commun. 2009, 389, 205–210. [Google Scholar] [CrossRef] [PubMed]
  62. Mauro, C.; Pacifico, F.; Lavorgna, A.; Mellone, S.; Iannetti, A.; Acquaviva, R.; Formisano, S.; Vito, P.; Leonardi, A. ABIN-1 binds to NEMO/IKKγ and co-operates with A20 in inhibiting NF-κB. J. Biol. Chem. 2006, 281, 18482–18488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Cruz, J.A.; Childs, E.E.; Amatya, N.; Garg, A.V.; Beyaert, R.; Kane, L.P.; Aneskievich, B.J.; Ma, A.; Gaffen, S.L. IL-17 Signaling Triggers Degradation of the Constitutive NF-κB Inhibitor ABIN-1. ImmunoHorizons 2017, 1, 133–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yang, B.; Kang, H.; Fung, A.; Zhao, H.; Wang, T.; Ma, D. The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis. Mediators Inflamm. 2014, 2014. [Google Scholar] [CrossRef] [PubMed]
  65. Wu, W.; He, K.; Guo, Q.; Chen, J.; Zhang, M.; Huang, K.; Yang, D.; Wu, L.; Deng, Y.; Luo, X.; et al. SSRP1 promotes colorectal cancer progression and is negatively regulated by miR-28-5p. J. Cell. Mol. Med. 2019, 23, 3118–3129. [Google Scholar] [CrossRef]
  66. Wang, C.; Wu, C.; Yang, Q.; Ding, M.; Zhong, J.; Zhang, C.Y.; Ge, J.; Wang, J.; Zhang, C. miR-28-5p acts as a tumor suppressor in renal cell carcinoma for multiple antitumor effects by targeting RAP1B. Oncotarget 2016, 7, 73888–73902. [Google Scholar] [CrossRef] [Green Version]
  67. Shi, X.; Teng, F. Down-regulated miR-28-5p in human hepatocellular carcinoma correlated with tumor proliferation and migration by targeting insulin-like growth factor-1 (IGF-1). Mol. Cell. Biochem. 2015, 408, 283–293. [Google Scholar] [CrossRef]
  68. Song, L.; Lin, C.; Gong, H.; Wang, C.; Liu, L.; Wu, J.; Tao, S.; Hu, B.; Cheng, S.Y.; Li, M.; et al. MiR-486 sustains NF-κB activity by disrupting multiple NF-κB-negative feedback loops. Cell Res. 2013, 23, 274–289. [Google Scholar] [CrossRef]
  69. Tan, G.; Wu, L.; Tan, J.; Zhang, B.; Tai, W.C.S.; Xiong, S.; Chen, W.; Yang, J.; Li, H. MIR-1180 promotes apoptotic resistance to human hepatocellular carcinoma via activation of NF-κB signaling pathway. Sci. Rep. 2016, 6, 1–11. [Google Scholar] [CrossRef] [Green Version]
  70. Xu, J.; Jiang, N.; Shi, H.; Zhao, S.; Yao, S.; Shen, H. MiR-28-5p promotes the development and progression of ovarian cancer through inhibition of N4BP1. Int. J. Oncol. 2017, 50, 1383–1391. [Google Scholar] [CrossRef] [Green Version]
  71. Zhu, G.; Wang, Z.; Mijiti, M.; Du, G.; Li, Y.; Dangmurenjiafu, G. MiR-28-5p promotes human glioblastoma cell growth through inactivation of FOXO1. Int. J. Clin. Exp. Pathol. 2019, 12, 2972–2980. [Google Scholar] [PubMed]
  72. Shembade, N.; Harhaj, E.W. Regulation of NF-κB signaling by the A20 deubiquitinase. Cell. Mol. Immunol. 2012, 9, 123–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Wertz, I.E.; Rourke, K.M.O.; Zhou, H.; Eby, M.; Aravind, L.; Seshagiri, S.; Wu, P.; Wiesmann, C.; Dixit, V.M. ligase domains of A20 downregulate NF- k B signalling. Nature 2004, 430, 1–6. [Google Scholar] [CrossRef]
  74. Shembade, N.; Harhaj, N.S.; Parvatiyar, K.; Copeland, N.G.; Jenkins, N.A.; Matesic, L.E.; Harhaj, E.W. The E3 ligase Itch negatively regulates inflammatory signaling pathways by controlling the function of the ubiquitin-editing enzyme A20. Nat. Immunol. 2008, 9, 254–262. [Google Scholar] [CrossRef]
  75. Ishihara, T.; Tsuda, H.; Hotta, A.; Kozaki, K.I.; Yoshida, A.; Noh, J.Y.; Ito, K.; Imoto, I.; Inazawa, J. ITCH is a putative target for a novel 20q11.22 amplification detected in anaplastic thyroid carcinoma cells by array-based comparative genomic hybridization. Cancer Sci. 2008, 99, 1940–1949. [Google Scholar] [CrossRef] [PubMed]
  76. Ho, K.C.; Zhou, Z.; She, Y.M.; Chun, A.; Cyr, T.D.; Yang, X. Itch E3 ubiquitin ligase regulates large tumor suppressor 1 tumor-suppressor stability. Proc. Natl. Acad. Sci. USA 2011, 108, 4870–4875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Salah, Z.; Melino, G.; Aqeilan, R.I. Negative regulation of the Hippo pathway by E3 ubiquitin ligase ITCH is sufficient to promote tumorigenicity. Cancer Res. 2011, 71, 2010–2020. [Google Scholar] [CrossRef] [Green Version]
  78. Salah, Z.; Itzhaki, E.; Aqeilan, R.I. The ubiquitin E3 ligase ITCH enhances breast tumor progression by inhibiting the Hippo tumor suppressor pathway. Oncotarget 2014, 5, 10886–10900. [Google Scholar] [CrossRef] [Green Version]
  79. Meng, J.; Tagalakis, A.D.; Hart, S.L. Silencing E3 Ubiqutin ligase ITCH as a potential therapy to enhance chemotherapy efficacy in p53 mutant neuroblastoma cells. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef]
  80. Rossi, M.; Rotblat, B.; Ansell, K.; Amelio, I.; Caraglia, M.; Misso, G.; Bernassola, F.; Cavasotto, C.N.; Knight, R.A.; Ciechanover, A.; et al. High throughput screening for inhibitors of the HECT ubiquitin E3 ligase ITCH identifies antidepressant drugs as regulators of autophagy. Cell Death Dis. 2014, 5, 1–12. [Google Scholar] [CrossRef] [Green Version]
  81. Liu, Y.M.; HuangFu, W.C.; Huang, H.L.; Wu, W.C.; Chen, Y.L.; Yen, Y.; Huang, H.L.; Nien, C.Y.; Lai, M.J.; Pan, S.L.; et al. 1,4-Naphthoquinones as inhibitors of Itch, a HECT domain-E3 ligase, and tumor growth suppressors in multiple myeloma. Eur. J. Med. Chem. 2017, 140, 84–91. [Google Scholar] [CrossRef] [PubMed]
  82. Zhu, J.; Bengtsson, B.O.; Mix, E.; Thorell, L.H.; Olsson, T.; Link, H. Effect of monoamine reuptake inhibiting antidepressants on major histocompatibility complex expression on macrophages in normal rats and rats with experimental allergic neuritis (EAN). Immunopharmacology 1994, 27, 225–244. [Google Scholar] [CrossRef]
  83. Kovalenko, A.; Chable-Bessia, C.; Cantarella, G.; Israël, A.; Wallach, D.; Courtois, G. The tumour suppressor CYLD negatively regulates NF-κB signalling by deubiquitination. Nature 2003, 424, 801–805. [Google Scholar] [CrossRef] [PubMed]
  84. Yin, J.; Weng, C.; Ma, J.; Chen, F.; Huang, Y.; Feng, M. MicroRNA-1288 promotes cell proliferation of human glioblastoma cells by repressing ubiquitin carboxyl-terminal hydrolase CYLD expression. Mol. Med. Rep. 2017, 16, 6764–6770. [Google Scholar] [CrossRef] [PubMed]
  85. Qiu, H.; Yuan, S.; Lu, X. miR-186 suppressed CYLD expression and promoted cell proliferation in human melanoma. Oncol. Lett. 2016, 12, 2301–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ni, F.; Zhao, H.; Cui, H.; Wu, Z.; Chen, L.; Hu, Z.; Guo, C.; Liu, Y.; Chen, Z.; Wang, X.; et al. MicroRNA-362-5p promotes tumor growth and metastasis by targeting CYLD in hepatocellular carcinoma. Cancer Lett. 2015, 356, 809–818. [Google Scholar] [CrossRef] [PubMed]
  87. Farshi, P.; Deshmukh, R.R.; Nwankwo, J.O.; Arkwright, R.T.; Cvek, B.; Liu, J.; Dou, Q.P. Deubiquitinases (DUBs) and DUB inhibitors: A patent review. Expert Opin. Ther. Pat. 2015, 25, 1191–1208. [Google Scholar] [CrossRef] [Green Version]
  88. Sun, S.C. CYLD: A tumor suppressor deubiquitinase regulating NF-B activation and diverse biological processes. Cell Death Differ. 2010, 17, 25–34. [Google Scholar] [CrossRef]
  89. Wang, J.H.; Wei, W.; Guo, Z.X.; Shi, M.; Guo, R. Decreased Cezanne expression is associated with the progression and poor prognosis in hepatocellular carcinoma. J. Transl. Med. 2015, 13, 1–10. [Google Scholar] [CrossRef] [Green Version]
  90. Tilborghs, S.; Corthouts, J.; Verhoeven, Y.; Arias, D.; Rolfo, C.; Trinh, X.B.; van Dam, P.A. The role of Nuclear Factor-kappa B signaling in human cervical cancer. Crit. Rev. Oncol. Hematol. 2017, 120, 141–150. [Google Scholar] [CrossRef]
  91. Musella, M.; Manic, G.; De Maria, R.; Vitale, I.; Sistigu, A. Type-I-interferons in infection and cancer: Unanticipated dynamics with therapeutic implications. Oncoimmunology 2017, 6, 1–12. [Google Scholar] [CrossRef] [Green Version]
  92. Castro, F.; Cardoso, A.P.; Gonçalves, R.M.; Serre, K.; Oliveira, M.J. Interferon-gamma at the crossroads of tumor immune surveillance or evasion. Front. Immunol. 2018, 9, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 2016, 375, 819–829. [Google Scholar] [CrossRef] [PubMed]
  94. Gao, J.; Shi, L.Z.; Zhao, H.; Chen, J.; Xiong, L.; He, Q.; Chen, T.; Roszik, J.; Bernatchez, C.; Woodman, S.E.; et al. Loss of IFN-γ Pathway Genes in Tumor Cells as a Mechanism of Resistance to Anti-CTLA-4 Therapy. Cell 2016, 167, 397–404.e9. [Google Scholar] [CrossRef] [PubMed]
  95. Stelloo, E.; Versluis, M.A.; Nijman, H.W.; De Bruyn, M.; Plat, A.; Osse, E.M.; Van Dijk, R.H.; Nout, R.A.; Creutzberg, C.L.; De Bock, G.H.; et al. Microsatellite instability derived JAK1 frameshift mutations are associated with tumor immune evasion in endometrioid endometrial cancer. Oncotarget 2016, 7, 39885–39893. [Google Scholar] [CrossRef] [Green Version]
  96. Smithy, J.W.; Moore, L.M.; Pelekanou, V.; Rehman, J.; Gaule, P.; Wong, P.F.; Neumeister, V.M.; Sznol, M.; Kluger, H.M.; Rimm, D.L. Nuclear IRF-1 expression as a mechanism to assess “Capability” to express PD-L1 and response to PD-1 therapy in metastatic melanoma. J. Immunother. Cancer 2017, 5, 1–9. [Google Scholar] [CrossRef] [Green Version]
  97. Bhat, M.Y.; Solanki, H.S.; Advani, J.; Khan, A.A.; Keshava Prasad, T.S.; Gowda, H.; Thiyagarajan, S.; Chatterjee, A. Comprehensive network map of interferon gamma signaling. J. Cell Commun. Signal. 2018, 12, 745–751. [Google Scholar] [CrossRef]
  98. Ivashkiv, L.B.; Donlin, L.T. Regulation of type i interferon responses. Nat. Rev. Immunol. 2014, 14, 36–49. [Google Scholar] [CrossRef] [Green Version]
  99. Propper, D.J.; Chao, D.; Braybrooke, J.P.; Bahl, P.; Thavasu, P.; Balkwill, F.; Turley, H.; Dobbs, N.; Gatter, K.; Talbot, D.C.; et al. Low-Dose IFN-γ Induces Tumor MHC Expression in Metastatic Malignant Melanoma. Clin. Cancer Res. 2003, 9, 84–92. [Google Scholar]
  100. Yang, I.; Kremen, T.J.; Giovannone, A.J.; Paik, E.; Odesa, S.K.; Prins, R.M.; Liau, L.M. Modulation of major histocompatibility complex Class I molecules and major histocompatibility complex-bound immunogenic peptides induced by interferon-α and interferon-γ treatment of human glioblastoma multiforme. J. Neurosurg. 2004, 100, 310–319. [Google Scholar] [CrossRef]
  101. George, P.M.; Badiger, R.; Alazawi, W.; Foster, G.R.; Mitchell, J.A. Pharmacology and therapeutic potential of interferons. Pharmacol. Ther. 2012, 135, 44–53. [Google Scholar] [CrossRef] [PubMed]
  102. Hemmerle, T.; Neri, D. The dose-dependent tumor targeting of antibody-IFNγ fusion proteins reveals an unexpected receptor-trapping mechanism in vivo. Cancer Immunol. Res. 2014, 2, 559–567. [Google Scholar] [CrossRef] [Green Version]
  103. Yang, Y.; Lim, O.; Kim, T.M.; Ahn, Y.O.; Choi, H.; Chung, H.; Min, B.; Her, J.H.; Cho, S.Y.; Keam, B.; et al. Phase I study of random healthy donor-derived allogeneic natural killer cell therapy in patients with malignant lymphoma or advanced solid tumors. Cancer Immunol. Res. 2016, 4, 215–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Cho, Y.-H.; Choi, M.G.; Kim, D.H.; Choi, Y.J.; Kim, S.Y.; Sung, K.J.; Lee, J.C.; Kim, S.-Y.; Rho, J.K.; Choi, C.M. Natural Killer Cells as a Potential Biomarker for Predicting Immunotherapy Efficacy in Patients with Non-Small Cell Lung Cancer. Target. Oncol. 2020, 15, 241–247. [Google Scholar] [CrossRef] [PubMed]
  105. Mazzaschi, G.; Facchinetti, F.; Missale, G.; Canetti, D.; Madeddu, D.; Zecca, A.; Veneziani, M.; Gelsomino, F.; Goldoni, M.; Buti, S.; et al. The circulating pool of functionally competent NK and CD8+ cells predicts the outcome of anti-PD1 treatment in advanced NSCLC. Lung Cancer 2019, 127, 153–163. [Google Scholar] [CrossRef] [PubMed]
  106. Castriconi, R.; Dondero, A.; Augugliaro, R.; Cantoni, C.; Carnemolla, B.; Sementa, A.R.; Negri, F.; Conte, R.; Corrias, M.V.; Moretta, L.; et al. Identification of 4Ig-B7-H3 as a neuroblastoma-associated molecule that exerts a protective role from an NK cell-mediated lysis. Proc. Natl. Acad. Sci. USA 2004, 101, 12640–12645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Lee, Y.H.; Martin-Orozco, N.; Zheng, P.; Li, J.; Zhang, P.; Tan, H.; Park, H.J.; Jeong, M.; Chang, S.H.; Kim, B.S.; et al. Inhibition of the B7-H3 immune checkpoint limits tumor growth by enhancing cytotoxic lymphocyte function. Cell Res. 2017, 27, 1034–1045. [Google Scholar] [CrossRef]
  108. Hu, Y.; Tian, Z.; Zhang, C. Natural Killer Cell-Based Immunotherapy for Cancer: Advances and Prospects. Engineering 2019, 5, 106–114. [Google Scholar] [CrossRef]
  109. Bernson, E.; Hallner, A.; E Sander, F.; Wilsson, O.; Werlenius, O.; Rydström, A.; Kiffin, R.; Brune, M.; Foà, R.; Aurelius, J.; et al. Impact of killer-immunoglobulin-like receptor and human leukocyte antigen genotypes on the efficacy of immunotherapy in acute myeloid leukemia. Leuk. 2017, 31, 2552–2559. [Google Scholar] [CrossRef] [Green Version]
  110. Boudreau, J.E.; Giglio, F.; Gooley, T.A.; Stevenson, P.A.; Le Luduec, J.-B.; Shaffer, B.C.; Rajalingam, R.; Hou, L.; Hurley, C.K.; Noreen, H.; et al. KIR3DL1/HLA-B Subtypes Govern Acute Myelogenous Leukemia Relapse After Hematopoietic Cell Transplantation. J. Clin. Oncol. 2017, 35, 2268–2278. [Google Scholar] [CrossRef]
  111. Lupo, K.B.; Matosevic, S. Natural Killer Cells as Allogeneic Effectors in Adoptive Cancer Immunotherapy. Cancers 2019, 11, 769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Dondero, A.; Pastorino, F.; Della Chiesa, M.; Corrias, M.V.; Morandi, F.; Pistoia, V.; Olive, D.; Bellora, F.; Locatelli, F.; Castellano, A.; et al. PD-L1 expression in metastatic neuroblastoma as an additional mechanism for limiting immune surveillance. Oncoimmunology 2016, 5, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Chen, S.; Crabill, G.A.; Pritchard, T.S.; McMiller, T.L.; Wei, P.; Pardoll, D.M.; Pan, F.; Topalian, S.L. Mechanisms regulating PD-L1 expression on tumor and immune cells. J. Immunother. Cancer 2019, 7, 1–12. [Google Scholar] [CrossRef] [PubMed]
  114. Chew, G.-L.; Campbell, A.E.; De Neef, E.; Sutliff, N.A.; Shadle, S.C.; Tapscott, S.J.; Bradley, R.K. DUX4 Suppresses MHC Class I to Promote Cancer Immune Evasion and Resistance to Checkpoint Blockade. Dev. Cell 2019, 50, 658–671.e7. [Google Scholar] [CrossRef]
  115. Bosnakovski, D.; da Silva, M.T.; Sunny, S.T.; Ener, E.T.; Toso, E.A.; Yuan, C.; Cui, Z.; Walters, M.A.; Jadhav, A.; Kyba, M. A novel P300 inhibitor reverses DUX4-mediated global histone H3 hyperacetylation, target gene expression, and cell death. Sci. Adv. 2019, 5, eaaw7781. [Google Scholar] [CrossRef] [Green Version]
  116. Lasko, L.M.; Jakob, C.G.; Edalji, R.P.; Qiu, W.; Montgomery, D.; Digiammarino, E.L.; Hansen, T.M.; Risi, R.M.; Frey, R.; Manaves, V.; et al. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature 2017, 550, 128–132. [Google Scholar] [CrossRef]
  117. Oliva, J.; Galasinski, S.; Richey, A.; Campbell, A.E.; Meyers, M.J.; Modi, N.; Zhong, J.W.; Tawil, R.; Tapscott, S.J.; Sverdrup, F.M. Clinically advanced p38 inhibitors suppress DUX4 expression in cellular and animal models of facioscapulohumeral muscular dystrophys. J. Pharmacol. Exp. Ther. 2019, 370, 219–230. [Google Scholar] [CrossRef] [PubMed]
  118. Yong, H.Y.; Koh, M.S.; Moon, A. The p38 MAPK inhibitors for the treatment of inflammatory diseases and cancer. Expert Opin. Investig. Drugs 2009, 18, 1893–1905. [Google Scholar] [CrossRef] [PubMed]
  119. Ding, L.W.; Sun, Q.Y.; Lin, D.C.; Chien, W.; Hattori, N.; Dong, X.M.; Gery, S.; Garg, M.; Doan, N.B.; Said, J.W.; et al. LNK (SH2B3): Paradoxical effects in ovarian cancer. Oncogene 2015, 34, 1463–1474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Ding, L.W.; Sun, Q.Y.; Edwards, J.J.; Fernández, L.T.; Ran, X.B.; Zhou, S.Q.; Scolyer, R.A.; Wilmott, J.S.; Thompson, J.F.; Doan, N.; et al. LNK suppresses interferon signaling in melanoma. Nat. Commun. 2019, 10. [Google Scholar] [CrossRef]
  121. Kriegel, A.J.; Baker, M.A.; Liu, Y.; Liu, P.; Cowley, A.W.; Liang, M. Endogenous MicroRNAs in human microvascular endothelial cells regulate mRNAs encoded by hypertension-related genes. Hypertension 2015, 66, 793–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Wang, Y.; Jin, B.J.; Chen, Q.; Yan, B.J.; Liu, Z.L. MicroRNA-29b upregulation improves myocardial fibrosis and cardiac function in myocardial infarction rats through targeting SH2B3. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10115–10122. [Google Scholar] [CrossRef]
  123. Saitoh, T.; Tun-Kyi, A.; Ryo, A.; Yamamoto, M.; Finn, G.; Fujita, T.; Akira, S.; Yamamoto, N.; Lu, K.P.; Yamaoka, S. Negative regulation of interferon-regulatory factor 3-dependent innate antiviral response by the prolyl isomerase Pin1. Nat. Immunol. 2006, 7, 598–605. [Google Scholar] [CrossRef]
  124. Chen, Y.; Wu, Y.; Yang, H.; Li, X.; Jie, M.; Hu, C.; Wu, Y.; Yang, S.; Yang, Y. Prolyl isomerase Pin1: A promoter of cancer and a target for therapy. Cell Death Dis. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  125. Hennig, L.; Christner, C.; Kipping, M.; Schelbert, B.; Rücknagel, K.P.; Grabley, S.; Küllertz, G.; Fischer, G. Selective inactivation of parvulin-like peptidyl-prolyl cis/trans isomerases by juglone. Biochemistry 1998, 37, 5953–5960. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, X.; Zhang, B.; Gao, J.; Wang, X.; Liu, Z. Regulation of the MicroRNA 200b (miRNA-200b) by transcriptional regulators PEA3 and ELK-1 protein affects expression of Pin1 protein to control anoikis. J. Biol. Chem. 2013, 288, 32742–32752. [Google Scholar] [CrossRef] [Green Version]
  127. Luo, M.L.; Gong, C.; Chen, C.H.; Lee, D.Y.; Hu, H.; Huang, P.; Yao, Y.; Guo, W.; Reinhardt, F.; Wulf, G.; et al. Prolyl isomerase pin1 acts downstream of mir200c to promote cancer stem-like cell traits in breast cancer. Cancer Res. 2014, 74, 3603–3616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Lee, K.H.; Lin, F.C.; Hsu, T.I.; Lin, J.T.; Guo, J.H.; Tsai, C.H.; Lee, Y.C.; Lee, Y.C.; Chen, C.L.; Hsiao, M.; et al. MicroRNA-296-5p (miR-296-5p) functions as a tumor suppressor in prostate cancer by directly targeting Pin1. Biochim. Biophys. Acta Mol. Cell Res. 2014, 1843, 2055–2066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Wei, S.; Kozono, S.; Kats, L.; Nechama, M.; Li, W.; Guarnerio, J.; Luo, M.; You, M.H.; Yao, Y.; Kondo, A.; et al. Active Pin1 is a key target of all-trans retinoic acid in acute promyelocytic leukemia and breast cancer. Nat. Med. 2015, 21, 457–466. [Google Scholar] [CrossRef] [PubMed]
  130. Campaner, E.; Rustighi, A.; Zannini, A.; Cristiani, A.; Piazza, S.; Ciani, Y.; Kalid, O.; Golan, G.; Baloglu, E.; Shacham, S.; et al. A covalent PIN1 inhibitor selectively targets cancer cells by a dual mechanism of action. Nat. Commun. 2017, 8. [Google Scholar] [CrossRef]
  131. Myers, M.P.; Andersen, J.N.; Cheng, A.; Tremblay, M.L.; Horvath, C.M.; Parisien, J.P.; Salmeen, A.; Barford, D.; Tonks, N.K. TYK2 and JAK2 Are Substrates of Protein-tyrosine Phosphatase 1B. J. Biol. Chem. 2001, 276, 47771–47774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. You, M.; Yu, D.-H.; Feng, G.-S. Shp-2 Tyrosine Phosphatase Functions as a Negative Regulator of the Interferon-Stimulated Jak/STAT Pathway. Mol. Cell. Biol. 1999, 19, 2416–2424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Bollu, L.R.; Mazumdar, A.; Savage, M.I.; Brown, P.H. Molecular pathways: Targeting protein tyrosine phosphatases in cancer. Clin. Cancer Res. 2017, 23, 2136–2142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Yi, T.; Pathak, M.K.; Lindner, D.J.; Ketterer, M.E.; Farver, C.; Borden, E.C. Anticancer Activity of Sodium Stibogluconate in Synergy with IFNs. J. Immunol. 2002, 169, 5978–5985. [Google Scholar] [CrossRef] [Green Version]
  135. Lantz, K.A.; Hart, S.G.E.; Planey, S.L.; Roitman, M.F.; Ruiz-White, I.A.; Wolfe, H.R.; McLane, M.P. Inhibition of PTP1B by trodusquemine (MSI-1436) causes fat-specific weight loss in diet-induced obese mice. Obesity 2010, 18, 1516–1523. [Google Scholar] [CrossRef]
  136. Huffaker, T.B.; Lee, S.H.; Tang, W.W.; Wallace, J.A.; Alexander, M.; Runtsch, M.C.; Larsen, D.K.; Thompson, J.; Ramstead, A.G.; Voth, W.P.; et al. Antitumor immunity is defective in T cell–specific microRNA-155– deficient mice and is rescued by immune checkpoint blockade. J. Biol. Chem. 2017, 292, 18530–18541. [Google Scholar] [CrossRef] [Green Version]
  137. Higgs, G.; Slack, F. The multiple roles of microRNA-155 in oncogenesis. J. Clin. Bioinforma. 2013, 3, 1–8. [Google Scholar] [CrossRef] [Green Version]
  138. Liu, B.; Mink, S.; Wong, K.A.; Stein, N.; Getman, C.; Dempsey, P.W.; Wu, H.; Shuai, K. PIAS1 selectively inhibits interferon-inducible genes and is important in innate immunity. Nat. Immunol. 2004, 5, 891–898. [Google Scholar] [CrossRef]
  139. Yuan, C.; Qi, J.; Zhao, X.; Gao, C. Smurf1 protein negatively regulates interferon-γ signaling through promoting STAT1 protein ubiquitination and degradation. J. Biol. Chem. 2012, 287, 17006–17015. [Google Scholar] [CrossRef] [Green Version]
  140. Liu, S.; Jiang, M.; Wang, W.; Liu, W.; Song, X.; Ma, Z.; Zhang, S.; Liu, L.; Liu, Y.; Cao, X. Nuclear RNF2 inhibits interferon function by promoting K33-linked STAT1 disassociation from DNA article. Nat. Immunol. 2018, 19, 41–50. [Google Scholar] [CrossRef]
  141. Sánchez-Beato, M.; Sánchez, E.; González-Carreró, J.; Morente, M.; Díez, A.; Sánchez-Verde, L.; Martín, M.C.; Cigudosa, J.C.; Vidal, M.; Piris, M.A. Variability in the expression of polycomb proteins in different normal and tumoral tissues. A pilot study using tissue microarrays. Mod. Pathol. 2006, 19, 684–694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Romero, I.; Martinez, M.; Garrido, C.; Collado, A.; Algarra, I.; Garrido, F.; Garcia-Lora, A.M. The tumour suppressor Fhit positively regulates MHC class I expression on cancer cells. J. Pathol. 2012, 227, 367–379. [Google Scholar] [CrossRef] [PubMed]
  143. Rai, K.; Akdemir, K.C.; Kwong, L.N.; Fiziev, P.; Wu, C.J.; Keung, E.Z.; Sharma, S.; Samant, N.S.; Williams, M.; Axelrad, J.B.; et al. Dual roles of RNF2 in melanoma progression. Cancer Discov. 2015, 5, 1314–1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Rabellino, A.; Andreani, C.; Scaglioni, P.P. The Role of PIAS SUMO E3-Ligases in Cancer. Cancer Res. 2017, 77, 1542–1547. [Google Scholar] [CrossRef] [Green Version]
  145. Yang, H.; Yu, N.; Xu, J.; Ding, X.; Deng, W.; Wu, G.; Li, X.; Hou, Y.; Liu, Z.; Zhao, Y.; et al. SMURF1 facilitates estrogen receptor a signaling in breast cancer cells. J. Exp. Clin. Cancer Res. 2018, 37, 1–12. [Google Scholar] [CrossRef] [PubMed]
  146. Chen, X.; Chen, S.; Li, Y.; Gao, Y.; Huang, S.; Li, H.; Zhu, Y. SMURF1-mediated ubiquitination of ARHGAP26 promotes ovarian cancer cell invasion and migration. Exp. Mol. Med. 2019, 51. [Google Scholar] [CrossRef] [Green Version]
  147. Nair, S.; Bist, P.; Dikshit, N.; Krishnan, M.N. Global functional profiling of human ubiquitome identifies E3 ubiquitin ligase DCST1 as a novel negative regulator of Type-I interferon signaling. Sci. Rep. 2016, 6, 1–13. [Google Scholar] [CrossRef] [Green Version]
  148. Ismail, I.H.; McDonald, D.; Strickfaden, H.; Xu, Z.; Hendzel, M.J. A small molecule inhibitor of polycomb repressive complex 1 inhibits ubiquitin signaling at DNA double-strand breaks. J. Biol. Chem. 2013, 288, 26944–26954. [Google Scholar] [CrossRef] [Green Version]
  149. Cao, Y.; Wang, C.; Zhang, X.; Xing, G.; Lu, K.; Gu, Y.; He, F.; Zhang, L. Selective small molecule compounds increase BMP-2 responsiveness by inhibiting Smurf1-mediated Smad1/5 degradation. Sci. Rep. 2014, 4, 1–11. [Google Scholar] [CrossRef] [Green Version]
  150. Zheng, H.; Qian, J.; Varghese, B.; Baker, D.P.; Fuchs, S. Ligand-Stimulated Downregulation of the Alpha Interferon Receptor: Role of Protein Kinase D2. Mol. Cell. Biol. 2011, 31, 710–720. [Google Scholar] [CrossRef] [Green Version]
  151. Azoitei, N.; Cobbaut, M.; Becher, A.; Van Lint, J.; Seufferlein, T. Protein kinase D2: A versatile player in cancer biology. Oncogene 2018, 37, 1263–1278. [Google Scholar] [CrossRef]
  152. Wei, N.; Chu, E.; Wipf, P.; Schmitz, J.C. Protein kinase D as a potential chemotherapeutic target for colorectal cancer. Mol. Cancer Ther. 2014, 13, 1130–1141. [Google Scholar] [CrossRef] [Green Version]
  153. Tandon, M.; Salamoun, J.M.; Carder, E.J.; Farber, E.; Xu, S.; Deng, F.; Tang, H.; Wipf, P.; Wang, Q.J. SD-208, a novel protein kinase D inhibitor, blocks prostate cancer cell proliferation and tumor Growth in Vivo by inducing G2/M cell cycle arrest. PLoS ONE 2015, 10, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Meissner, T.B.; Li, A.; Biswas, A.; Lee, K.H.; Liu, Y.J.; Bayir, E.; Iliopoulos, D.; Van Den Elsen, P.J.; Kobayashi, K.S. NLR family member NLRC5 is a transcriptional regulator of MHC class I genes. Proc. Natl. Acad. Sci. USA 2010, 107, 13794–13799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Staehli, F.; Ludigs, K.; Heinz, L.X.; Seguín-Estévez, Q.; Ferrero, I.; Braun, M.; Schroder, K.; Rebsamen, M.; Tardivel, A.; Mattmann, C.; et al. NLRC5 Deficiency Selectively Impairs MHC Class I- Dependent Lymphocyte Killing by Cytotoxic T Cells. J. Immunol. 2012, 188, 3820–3828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Kuenzel, S.; Till, A.; Winkler, M.; Häsler, R.; Lipinski, S.; Jung, S.; Grötzinger, J.; Fickenscher, H.; Schreiber, S.; Rosenstiel, P. The Nucleotide-Binding Oligomerization Domain-Like Receptor NLRC5 Is Involved in IFN-Dependent Antiviral Immune Responses. J. Immunol. 2010, 184, 1990–2000. [Google Scholar] [CrossRef]
  157. Kobayashi, K.S.; Van Den Elsen, P.J. NLRC5: A key regulator of MHC class I-dependent immune responses. Nat. Rev. Immunol. 2012, 12, 813–820. [Google Scholar] [CrossRef]
  158. Ozcan, M.; Janikovits, J.; von Knebel Doeberitz, M.; Kloor, M. Complex pattern of immune evasion in MSI colorectal cancer. Oncoimmunology 2018, 7, 1–10. [Google Scholar] [CrossRef] [Green Version]
  159. Yoshihama, S.; Roszik, J.; Downs, I.; Meissner, T.B.; Vijayan, S.; Chapuy, B.; Sidiq, T.; Shipp, M.A.; Lizee, G.A.; Kobayashi, K.S. NLRC5/MHC class I transactivator is a target for immune evasion in cancer. Proc. Natl. Acad. Sci. USA 2016, 113, 5999–6004. [Google Scholar] [CrossRef] [Green Version]
  160. Rodriguez, G.M.; Bobbala, D.; Serrano, D.; Mayhue, M.; Champagne, A.; Saucier, C.; Steimle, V.; Kufer, T.A.; Menendez, A.; Ramanathan, S.; et al. NLRC5 elicits antitumor immunity by enhancing processing and presentation of tumor antigens to CD8+ T lymphocytes. Oncoimmunology 2016, 5, 1–12. [Google Scholar] [CrossRef] [Green Version]
  161. Tang, F.; Xu, Y.; Zhao, B. NLRC5: New cancer buster? Mol. Biol. Rep. 2020, 47, 2265–2277. [Google Scholar] [CrossRef] [PubMed]
  162. Huynh, J.; Chand, A.; Gough, D.; Ernst, M. Therapeutically exploiting STAT3 activity in cancer—using tissue repair as a road map. Nat. Rev. Cancer 2019, 19, 82–96. [Google Scholar] [CrossRef]
  163. Blaskovich, M.A.; Sun, J.; Cantor, A.; Turkson, J.; Jove, R.; Sebti, S.M. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 2003, 63, 1270–1279. [Google Scholar]
  164. Nam, S.; Buettner, R.; Turkson, J.; Kim, D.; Cheng, J.Q.; Muehlbeyer, S.; Hippe, F.; Vatter, S.; Merz, K.H.; Eisenbrand, G.; et al. Indirubin derivatives inhibit Stat3 signaling and induce apoptosis in human cancer cells. Proc. Natl. Acad. Sci. USA 2005, 102, 5998–6003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Kotha, A.; Sekharam, M.; Cilenti, L.; Siddiquee, K.; Khaled, A.; Zervos, A.S.; Carter, B.; Turkson, J.; Jove, R. Resveratrol inhibits Src and Stat3 signaling and induces the apoptosis of malignant cells containing activated Stat3 protein. Mol. Cancer Ther. 2006, 5, 621–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Schust, J.; Sperl, B.; Hollis, A.; Mayer, T.U.; Berg, T. Stattic: A Small-Molecule Inhibitor of STAT3 Activation and Dimerization. Chem. Biol. 2006, 13, 1235–1242. [Google Scholar] [CrossRef] [Green Version]
  167. Yang, F.; Van Meter, T.E.; Buettner, R.; Hedvat, M.; Liang, W.; Kowolik, C.M.; Mepani, N.; Mirosevich, J.; Nam, S.; Chen, M.Y.; et al. Sorafenib inhibits signal transducer and activator of transcription 3 signaling associated with growth arrest and apoptosis of medulloblastomas. Mol. Cancer Ther. 2008, 7, 3519–3526. [Google Scholar] [CrossRef] [Green Version]
  168. Xin, H.; Zhang, C.; Herrmann, A.; Du, Y.; Figlin, R.; Yu, H. Sunitinib inhibition of Stat3 induces renal cell carcinoma tumor cell apoptosis and reduces immunosuppressive cells. Cancer Res. 2009, 69, 2506–2513. [Google Scholar] [CrossRef] [Green Version]
  169. McFarland, B.C.; Gray, G.K.; Nozell, S.E.; Hong, S.W.; Benveniste, E.N. Activation of the NF-κB pathway by the STAT3 inhibitor JSI-124 in human glioblastoma cells. Mol. Cancer Res. 2013, 11, 494–505. [Google Scholar] [CrossRef] [Green Version]
  170. Zhu, Y.; Ye, T.; Yu, X.; Lei, Q.; Yang, F.; Xia, Y.; Song, X.; Liu, L.; Deng, H.; Gao, T.; et al. Nifuroxazide exerts potent anti-tumor and anti-metastasis activity in melanoma. Sci. Rep. 2016, 6, 1–13. [Google Scholar] [CrossRef] [Green Version]
  171. Yang, H.; Yamazaki, T.; Pietrocola, F.; Zhou, H.; Zitvogel, L.; Ma, Y.; Kroemer, G. STAT3 inhibition enhances the therapeutic efficacy of immunogenic chemotherapy by stimulating type 1 interferon production by cancer cells. Cancer Res. 2015, 75, 3812–3822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Lu, C.; Talukder, A.; Savage, N.M.; Singh, N.; Liu, K. JAK-STAT-mediated chronic inflammation impairs cytotoxic T lymphocyte activation to decrease anti-PD-1 immunotherapy efficacy in pancreatic cancer. Oncoimmunology 2017, 6, 1–15. [Google Scholar] [CrossRef] [Green Version]
  173. Cohen, E.E.W.; Harrington, K.J.; Hong, D.S.; Mesia, R.; Brana, I.; Perez Segura, P.; Wise-Draper, T.; Scott, M.L.; Mitchell, P.D.; Mugundu, G.M.; et al. A phase Ib/II study (SCORES) of durvalumab (D) plus danvatirsen (DAN.; AZD9150) or AZD5069 (CX2i) in advanced solid malignancies and recurrent/metastatic head and neck squamous cell carcinoma (RM-HNSCC): Updated results. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, viii372. [Google Scholar] [CrossRef]
  174. D’amico, S.; Shi, J.; Martin, B.L.; Crawford, H.C.; Petrenko, O.; Reich, N.C. STAT3 is a master regulator of epithelial identity and KRAS-driven tumorigenesis. Genes Dev. 2018, 32, 1175–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Barber, G.N. STING: Infection, inflammation and cancer. Nat. Rev. Immunol. 2015, 15, 760–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Abe, T.; Barber, G.N. Cytosolic-DNA-Mediated, STING-Dependent Proinflammatory Gene Induction Necessitates Canonical NF- B Activation through TBK1. J. Virol. 2014, 88, 5328–5341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Challa, S.V.; Zhou, S.; Sheri, A.; Padmanabhan, S.; Meher, G.; Gimi, R.; Schmidt, D.; Cleary, D.; Afdhal, N.; Iyer, R. Preclinical studies of SB 11285, a novel STING agonist for immuno-oncology. J. Clin. Oncol. 2017, 35, e14616. [Google Scholar] [CrossRef]
  178. Wang-Bishop, L.; Wehbe, M.; Shae, D.; James, J.; Hacker, B.C.; Garland, K.; Chistov, P.P.; Rafat, M.; Balko, J.M.; Wilson, J.T. Potent STING activation stimulates immunogenic cell death to enhance antitumor immunity in neuroblastoma. J. Immunother. Cancer 2020, 8, 1–17. [Google Scholar] [CrossRef] [Green Version]
  179. Ager, C.R.; Reilley, M.J.; Nicholas, C.; Bartkowiak, T.; Jaiswal, A.R.; Curran, M.A. Intratumoral STING activation with T-cell checkpoint modulation generates systemic antitumor immunity. Cancer Immunol. Res. 2017, 5, 676–684. [Google Scholar] [CrossRef] [Green Version]
  180. Ghaffari, A.; Peterson, N.; Khalaj, K.; Vitkin, N.; Robinson, A.; Francis, J.A.; Koti, M. Sting agonist therapy in combination with pd-1 immune checkpoint blockade enhances response to carboplatin chemotherapy in high-grade serous ovarian cancer. Br. J. Cancer 2018, 119, 440–449. [Google Scholar] [CrossRef]
  181. Meric-Bernstam, F.; Sandhu, S.K.; Hamid, O.; Spreafico, A.; Kasper, S.; Dummer, R.; Shimizu, T.; Steeghs, N.; Lewis, N.; Talluto, C.C.; et al. Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas. J. Clin. Oncol. 2019, 37. [Google Scholar] [CrossRef]
  182. Seliger, B.; Harders, C.; Lohmann, S.; Momburg, F.; Urlinger, S.; Tampé, R.; Huber, C. Down-regulation of the MHC class I antigen-processing machinery after oncogenic transformation of murine fibroblasts. Eur. J. Immunol. 1998, 28, 122–133. [Google Scholar] [CrossRef]
  183. Herrmann, F.; Lehr, H.A.; Drexler, I.; Sutter, G.; Hengstler, J.; Wollscheid, U.; Seliger, B. HER-2/neu-Mediated Regulation of Components of the MHC Class I Antigen-Processing Pathway. Cancer Res. 2004, 64, 215–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Bradley, S.D.; Chen, Z.; Melendez, B.; Talukder, A.; Khalili, J.S.; Rodriguez-Cruz, T.; Liu, S.; Whittington, M.; Deng, W.; Li, F.; et al. BRAFV600E co-opts a conserved MHC class I internalization pathway to diminish antigen presentation and CD8+ T-cell recognition of melanoma. Cancer Immunol. Res. 2015, 3, 602–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Brea, E.J.; Oh, C.Y.; Manchado, E.; Budhu, S.; Gejman, R.S.; Mo, G.; Mondello, P.; Han, J.E.; Jarvis, C.A.; Ulmert, D.; et al. Kinase regulation of human MHC class i molecule expression on cancer cells. Cancer Immunol. Res. 2016, 4, 936–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Versteeg, R.; Noordermeer, I.A.; Krüse-Wolters, M.; Ruiter, D.J.; Schrier, P.I. c-myc down-regulates class I HLA expression in human melanomas. EMBO J. 1988, 7, 1023–1029. [Google Scholar] [CrossRef]
  187. Yang, W.; Li, Y.; Gao, R.; Xiu, Z.; Sun, T. MHC class I dysfunction of glioma stem cells escapes from CTL-mediated immune response via activation of Wnt/β-catenin signaling pathway. Oncogene 2020, 39, 1098–1111. [Google Scholar] [CrossRef]
  188. Lulli, D.; Carbone, M.L.; Pastore, S. The MEK inhibitors trametinib and cobimetinib induce a type I interferon response in human keratinocytes. Int. J. Mol. Sci. 2017, 18, 2227. [Google Scholar] [CrossRef] [Green Version]
  189. Watanabe, S.; Hayashi, H.; Haratani, K.; Shimizu, S.; Tanizaki, J.; Sakai, K.; Kawakami, H.; Yonesaka, K.; Tsurutani, J.; Togashi, Y.; et al. Mutational activation of the epidermal growth factor receptor down-regulates major histocompatibility complex class I expression via the extracellular signal-regulated kinase in non–small cell lung cancer. Cancer Sci. 2019, 110, 52–60. [Google Scholar] [CrossRef] [Green Version]
  190. Sapkota, B.; Hill, C.E.; Pollack, B.P. Vemurafenib enhances MHC induction in BRAFV600E homozygous melanoma cells. Oncoimmunology 2013, 2, e22890. [Google Scholar] [CrossRef] [Green Version]
  191. Mimura, K.; Ando, T.; Poschke, I.; Mougiakakos, D.; Johansson, C.C.; Ichikawa, J.; Okita, R.; Nishimura, M.I.; Handke, D.; Krug, N.; et al. T cell recognition of HLA-A2 restricted tumor antigens is impaired by the oncogene HER2. Int. J. Cancer 2011, 128, 390–401. [Google Scholar] [CrossRef] [PubMed]
  192. Maruyama, T.; Mimura, K.; Sato, E.; Watanabe, M.; Mizukami, Y.; Kawaguchi, Y.; Ando, T.; Kinouchi, H.; Fujii, H.; Kono, K. Inverse correlation of HER2 with MHC class i expression on oesophageal squamous cell carcinoma. Br. J. Cancer 2010, 103, 552–559. [Google Scholar] [CrossRef] [Green Version]
  193. Hastings, K.; Yu, H.A.; Wei, W.; Sanchez-Vega, F.; Deveaux, M.; Choi, J.; Rizvi, H.; Lisberg, A.; Truini, A.; Lydon, C.A.; et al. EGFR mutation subtypes and response to immune checkpoint blockade treatment in non-small-cell lung cancer. Ann. Oncol. 2019, 30, 1311–1320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Zage, P.E.; Sirisaengtaksin, N.; Liu, Y.; Gireud, M.; Brown, B.S.; Palla, S.; Richards, K.N.; Hughes, D.P.M.; Bean, A.J. UBE4B levels are correlated with clinical outcomes in neuroblastoma patients and with altered neuroblastoma cell proliferation and sensitivity to epidermal growth factor receptor inhibitors. Cancer 2013, 119, 915–923. [Google Scholar] [CrossRef] [PubMed]
  195. Leibowitz, M.S.; Srivastava, R.M.; Filho, P.A.A.; Egloff, A.M.; Wang, L.; Seethala, R.R.; Ferrone, S.; Ferris, R.L. SHP2 is overexpressed and inhibits pSTAT1-mediated APM component expression, T cell attracting chemokine secretion, and CTL recognition in head and neck cancer cells. Clin. Cancer Res. 2013, 19, 798–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Agazie, Y.M.; Hayman, M.J. Molecular Mechanism for a Role of SHP2 in Epidermal Growth Factor Receptor Signaling. Mol. Cell. Biol. 2003, 23, 7875–7886. [Google Scholar] [CrossRef] [Green Version]
  197. Grandis, J.R.; Drenning, S.D.; Zeng, Q.; Watkins, S.C.; Melhem, M.F.; Endo, S.; Johnson, D.E.; Huang, L.; He, Y.; Kim, J.D. Constitutive activation of stat3 signaling abrogates apoptosis in squamous cell carcinogenesis in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 4227–4232. [Google Scholar] [CrossRef] [Green Version]
  198. Garrido, G.; Rabasa, A.; Garrido, C.; Chao, L.; Garrido, F.; García-Lora, Á.M.; Sánchez-Ramírez, B. Upregulation of HLA Class I Expression on Tumor Cells by the Anti-EGFR Antibody Nimotuzumab. Front. Pharmacol. 2017, 8, 1–11. [Google Scholar] [CrossRef] [Green Version]
  199. Srivastava, R.M.; Trivedi, S.; Concha-Benavente, F.; Hyun-Bae, J.; Wang, L.; Seethala, R.R.; Branstetter IV, B.F.; Ferrone, S.; Ferris, R.L. Stat1-induced HLA class i upregulation enhances immunogenicity and clinical response to anti-EGFR mab cetuximab therapy in HNC patients. Cancer Immunol. Res. 2015, 3, 936–945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Pollack, B.P.; Sapkota, B.; Cartee, T.V. Epidermal growth factor receptor inhibition augments the expression of MHC class I and II genes. Clin. Cancer Res. 2011, 17, 4400–4413. [Google Scholar] [CrossRef] [Green Version]
  201. Freeman, A.J.; Vervoort, S.J.; Ramsbottom, K.M.; Kelly, M.J.; Michie, J.; Pijpers, L.; Johnstone, R.W.; Kearney, C.J.; Oliaro, J. Natural Killer Cells Suppress T Cell-Associated Tumor Immune Evasion. Cell Rep. 2019, 28, 2784–2794.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. He, S.; Yin, T.; Li, D.; Gao, X.; Wan, Y.; Ma, X.; Ye, T.; Guo, F.; Sun, J.; Lin, Z.; et al. Enhanced interaction between natural killer cells and lung cancer cells: Involvement in gefitinib-mediated immunoregulation. J. Transl. Med. 2013, 11, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Van’t Veer, L.J.; Beijersbergen, R.L.; Bernards, R. N-myc suppresses major histocompatibility complex class I gene expression through down-regulation of the p50 subunit of NF-kappa B. EMBO J. 1993, 12, 195–200. [Google Scholar] [CrossRef] [Green Version]
  204. Forloni, M.; Albini, S.; Limongi, M.Z.; Cifaldi, L.; Boldrini, R.; Nicotra, M.R.; Giannini, G.; Natali, P.G.; Giacomini, P.; Fruci, D. NF-κB, and not MYCN, regulates MHC class I and endoplasmic reticulum aminopeptidases in human neuroblastoma cells. Cancer Res. 2010, 70, 916–924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Hodge, J.W.; Garnett, C.T.; Farsaci, B.; Palena, C.; Tsang, K.Y.; Ferrone, S.; Gameiro, S.R. Chemotherapy-induced immunogenic modulation of tumor cells enhances killing by cytotoxic T lymphocytes and is distinct from immunogenic cell death. Int. J. Cancer 2013, 133, 624–636. [Google Scholar] [CrossRef] [PubMed]
  206. Khallouf, H.; Märten, A.; Serba, S.; Teichgräber, V.; Büchler, M.W.; Jäger, D.; Schmidt, J. 5-fluorouracil and interferon-α immunochemotherapy enhances immunogenicity of murine pancreatic cancer through upregulation of NKG2D ligands and MHC class i. J. Immunother. 2012, 35, 245–253. [Google Scholar] [CrossRef]
  207. Wan, S.; Pestka, S.; Jubin, R.G.; Lyu, Y.L.; Tsai, Y.C.; Liu, L.F. Chemotherapeutics and radiation stimulate MHC class i expression through elevated interferon-beta signaling in breast cancer cells. PLoS ONE 2012, 7, e32542. [Google Scholar] [CrossRef]
  208. Iwai, T.; Sugimoto, M.; Wakita, D.; Yorozu, K.; Kurasawa, M.; Yamamoto, K. Topoisomerase I inhibitor, irinotecan, depletes regulatory T cells and up-regulates MHC class I and PD-L1 expression, resulting in a supra-additive antitumor effect when combined with anti- PD-L1 antibodies. Oncotarget 2018, 9, 31411–31421. [Google Scholar] [CrossRef]
  209. Alagkiozidis, I.; Facciabene, A.; Tsiatas, M.; Carpenito, C.; Benencia, F.; Adams, S.; Jonak, Z.; June, C.H.; Powell, D.J.; Coukos, G. Time-dependent cytotoxic drugs selectively cooperate with IL-18 for cancer chemo-immunotherapy. J. Transl. Med. 2011, 9, 77. [Google Scholar] [CrossRef] [Green Version]
  210. De Mora-García, M.L.; Duenas-González, A.; Hernández-Montes, J.; De la Cruz-Hernández, E.; Pérez-Cárdenas, E.; Weiss-Steider, B.; Santiago-Osorio, E.; Ortíz-Navarrete, V.F.; Rosales, V.H.; Cantú, D.; et al. Up-regulation of HLA class-I antigen expression and antigen-specific CTL response in cervical cancer cells by the demethylating agent hydralazine and the histone deacetylase inhibitor valproic acid. J. Transl. Med. 2006, 4, 1–14. [Google Scholar] [CrossRef] [Green Version]
  211. Khan, A.N.H.; Gregorie, C.J.; Tomasi, T.B. Histone deacetylase inhibitors induce TAP, LMP, Tapasin genes and MHC class I antigen presentation by melanoma cells. Cancer Immunol. Immunother. 2008, 57, 647–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Ritter, C.; Fan, K.; Paschen, A.; Reker Hardrup, S.; Ferrone, S.; Nghiem, P.; Ugurel, S.; Schrama, D.; Becker, J.C. Epigenetic priming restores the HLA class-I antigen processing machinery expression in Merkel cell carcinoma. Sci. Rep. 2017, 7, 1–11. [Google Scholar] [CrossRef] [Green Version]
  213. Setiadi, A.F.; Omilusik, K.; David, M.D.; Seipp, R.P.; Hartikainen, J.; Gopaul, R.; Choi, K.B.; Jefferies, W.A. Epigenetic enhancement of antigen processing and presentation promotes immune recognition of tumors. Cancer Res. 2008, 68, 9601–9607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Suraweera, A.; O’Byrne, K.J.; Richard, D.J. Combination therapy with histone deacetylase inhibitors (HDACi) for the treatment of cancer: Achieving the full therapeutic potential of HDACi. Front. Oncol. 2018, 8, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Serrano, A.; Tanzarella, S.; Lionello, I.; Mendez, R.; Traversari, C.; Ruiz-Cabello, F.; Garrido, F. Expression of HLA class I antigens and restoration of antigen-specific CTL response in melanoma cells following 5-aza-2′-deoxycytidine treatment. Int. J. Cancer 2001, 94, 243–251. [Google Scholar] [CrossRef]
  216. Luo, N.; Nixon, M.J.; Gonzalez-Ericsson, P.I.; Sanchez, V.; Opalenik, S.R.; Li, H.; Zahnow, C.A.; Nickels, M.L.; Liu, F.; Tantawy, M.N.; et al. DNA methyltransferase inhibition upregulates MHC-I to potentiate cytotoxic T lymphocyte responses in breast cancer. Nat. Commun. 2018, 9, 248. [Google Scholar] [CrossRef]
  217. Fonsatti, E.; Nicolay, H.J.M.; Sigalotti, L.; Calabrò, L.; Pezzani, L.; Colizzi, F.; Altomonte, M.; Guidoboni, M.; Marincola, F.M.; Maio, M. Functional up-regulation of human leukocyte antigen class I antigens expression by 5-aza-2′-deoxycytidine in cutaneous melanoma: Immunotherapeutic implications. Clin. Cancer Res. 2007, 13, 3333–3338. [Google Scholar] [CrossRef] [Green Version]
  218. Santin, A.D.; Hermonat, P.L.; Ravaggi, A.; Chiriva-Internati, M.; Pecorelli, S.; Parham, G.P. Retinoic acid up-regulates the expression of major histocompatibility complex molecules and adhesion/costimulation molecules (specifically, intercellular adhesion molecule ICAM-1) in human cervical cancer. In Proceedings of the American Journal of Obstetrics and Gynecology; Mosby Inc.: St. Louis, MO, USA, 1998; Volume 179, pp. 1020–1025. [Google Scholar]
  219. Castriconi, R.; Dondero, A.; Cilli, M.; Ognio, E.; Pezzolo, A.; De Giovanni, B.; Gambini, C.; Pistoia, V.; Moretta, L.; Moretta, A.; et al. Human NK cell infusions prolong survival of metastatic human neuroblastoma-bearing NOD/scid mice. Cancer Immunol. Immunother. 2007, 56, 1733–1742. [Google Scholar] [CrossRef]
Figure 1. Major histocompatibility complex class I (MHC-I) antigen processing and presentation is a complex, multi-step process and can be dysregulated in cancer at multiple levels. APM = antigen processing machinery; ERAP = ER aminopeptidases; MHC-I = major histocompatibility complex I; TCR = T-cell receptor
Figure 1. Major histocompatibility complex class I (MHC-I) antigen processing and presentation is a complex, multi-step process and can be dysregulated in cancer at multiple levels. APM = antigen processing machinery; ERAP = ER aminopeptidases; MHC-I = major histocompatibility complex I; TCR = T-cell receptor
Cancers 12 01760 g001
Figure 2. NFkB-induced expression of MHC-I. Transcriptional activation of MHC-I heavy chain, but also other genes encoding for antigen processing machinery (APM) proteins, can be initiated by both the canonical (A) and non-canonical (B) NFkB pathway. IkB = inhibitors of kB; IKK = IkB kinase; NEMO = NF-kappa-B essential modulator; NIK = NFkB-inducing kinase; RIP-1 = receptor-interacting protein 1; TAK1 = TGF-β-activated kinase 1; TLR = toll-like receptor; TNFR = TNFα receptor; TRAF = TNF receptor-associated factor.
Figure 2. NFkB-induced expression of MHC-I. Transcriptional activation of MHC-I heavy chain, but also other genes encoding for antigen processing machinery (APM) proteins, can be initiated by both the canonical (A) and non-canonical (B) NFkB pathway. IkB = inhibitors of kB; IKK = IkB kinase; NEMO = NF-kappa-B essential modulator; NIK = NFkB-inducing kinase; RIP-1 = receptor-interacting protein 1; TAK1 = TGF-β-activated kinase 1; TLR = toll-like receptor; TNFR = TNFα receptor; TRAF = TNF receptor-associated factor.
Cancers 12 01760 g002
Figure 3. Interferon (IFN)-induced expression of MHC-I. Transcriptional activation of MHC-I heavy chain, but also other genes encoding for antigen processing machinery (APM) proteins, can be initiated by both the type II (A) and I (B) IFN pathway. GAS = gamma-activated site; IFNAR = IFNα receptor; IFNGR = IFNγ receptor; ISGF3 = IFN-stimulated gene factor 3; IRF = interferon regulatory factor; ISRE = interferon-stimulated response element; JAK = Janus Activated Kinase; STAT = signal transducer and activator of transcription; TYK2 = tyrosine kinase 2.
Figure 3. Interferon (IFN)-induced expression of MHC-I. Transcriptional activation of MHC-I heavy chain, but also other genes encoding for antigen processing machinery (APM) proteins, can be initiated by both the type II (A) and I (B) IFN pathway. GAS = gamma-activated site; IFNAR = IFNα receptor; IFNGR = IFNγ receptor; ISGF3 = IFN-stimulated gene factor 3; IRF = interferon regulatory factor; ISRE = interferon-stimulated response element; JAK = Janus Activated Kinase; STAT = signal transducer and activator of transcription; TYK2 = tyrosine kinase 2.
Cancers 12 01760 g003
Figure 4. Potential therapeutic interference to boost MHC-I antigen presentation of tumors. Targetable negative pathway regulators are shown in red, (general groups of) compounds positively affecting pathway activation are shown in green.
Figure 4. Potential therapeutic interference to boost MHC-I antigen presentation of tumors. Targetable negative pathway regulators are shown in red, (general groups of) compounds positively affecting pathway activation are shown in green.
Cancers 12 01760 g004
Table 1. Overview of described pathways and potential therapeutic strategies to boost MHC-I antigen presentation.
Table 1. Overview of described pathways and potential therapeutic strategies to boost MHC-I antigen presentation.
RegulationPathwayEffectorDescribed Mechanism(s)Potential Therapeutic StrategyClinical Status
Negative RegulatorsNFkB pathwayN4BP11. Cezanne-1 stabilization
2. Preventing recruitment of NEMO to RIP1
miRNA-28-5p [65,66,67,70,71]Pre-clinical
Cezanne-1Deubiquitination and stabilization of TRAF3Aspecific DUB inhibitors (e.g., Ubal) [87]
miR-1180 [69], miR-486 [68]
Cyanopyrrolidine derivates (WO2017109488)
Pre-clinical
TNIP11. Preventing recruitment of NEMO to RIP1
2. Preventing degradation of the IkB p105 into p50
3. A20 stabilization
IL-17 [63]
miR-1180 [69], miR-486 [68]
Pre-clinical
A201. Deubiquitination of NEMO
2. Deubiquitination of RIP-1 and TRAF6
3. Degradation of RIP-1
Aspecific DUB inhibitors (e.g., Ubal) [87]
miR-1180 [69], miR-486 [68]
Pre-clinical
TAX1BP1Itch recruitment to A20
ItchControlling interaction between A20 and RIP1/TRAF6Clomipramine and norclomipramine [80]
1,4-naphthoquinone 10E [81]
FDA-approved antidepressants
Pre-clinical
CYLD1. Deubiquitination of NEMO
2. Deubiquitination of TRAF2
miR-1288 [84], miR-186 [85], miR-362-5p [86]Pre-clinical
Type I/II IFN pathwayDUX41. JAK 1/2 downregulation
2. STAT2 downregulation
p300 inhibitors [115]
p38 inhibitors [117]
Pre-clinical
Several Phase II Trials (Rheumatoid Arthritis, Asthma, LMNA-related cardiomyopathy)
LNK (SH2B3)Dephosphorylation of STAT1miR-29b [122], miR-30-5p [121], miR-98 [121], miR181a-5p [121]
PTPN1 (PTP1B)1. Dephosphorylation of TYK2
2. Dephosphorylation of JAK2
MSI-1436C [135]Phase I Trial, metastatic breast cancer
PTPN2 (TC-PTP)1. Dephosphorylation of JAK1miRNA-155 [136]Pre-clinical
PTPN11 (SHP2)1. Dephosphorylation of JAK1
2. Dephosphorylation of STAT1
3. Dephosphorylation of STAT2
Sodium stibogluconate [134]Phase II Trial, Leishmaniasis, stage IV melanoma, advanced solid tumor
RNF2Polyubiquitination of STAT1, resulting in release from DNATrametinib [143]
PRT4165 [148]
FDA-approved in several advanced-stage cancers
Pre-clinical
Smurf1Degradation of STAT1A01 [149]Pre-clinical
PIASInhibiting STAT1 promotor recruitment
Type I IFN pathwayDCST1Ubiquitination and degradation of STAT2
PKD2Stimulating ubiquitination and endocytosis of IFNAR1CRT0066101 [152]
SD-208 [153]
Pre-clinical
Pin1Ubiquitination and degradation of IRF3miR-200b [126], miR-200c [127], miR-296-5p [128]
ATRA [129]
KPT-6566 [130]
Juglone [125]
Pre-clinical
FDA-approved in cancer
Pre-clinical
Pre-clinical
NFkB and type I/II IFN pathwaySTAT31. Negative feedback loop of pro-inflammatory type I signaling
2. Inhibition of IKK activation
STAT3 inhibitors:
JSI-124 [169]
Indirubin [164]
Resveratrol [165]
Nifuroxazide [170]
Static [166]
Sorafenib [167]
Sunitinib [168]
Pre-clinical
Phase 3/4 trials in acute promyelocytic leukemia, phase 2/3 in dermatitis and psoriasis
Phase 2/3 in congestive heart failure, Friedreich Ataxia, Gulf War Illness, Lymphangioleiomyomatosis, and infertility
FDA-approved antibiotic
Pre-clinical
Pre-clinical
Pre-clinical
EGFR receptor family (including HER2/neu)1. STAT1 inactivation
2. Stimulation of MAPK pathway
3. STAT3 activation
4. Fhit degradation
Tyrosine Kinase inhibitors:
EGFR inhibitors (e.g., nimotuzumab [198], cetuximab [199], afatinib [185], erlotinib [185], gefitinib [202]).
FDA-approved in cancer
NMYCInhibition of p50 (although inconsistent)
MAPK pathway1. Decreases IRF1 activity
2. Decreases STAT1 expression
Tyrosine Kinase inhibitors:
MEK-inhibitors (e.g., trametinib [188,189], cobimetinib [188], selumetinib [2])
BRAF inhibitors (e.g., vemurafenib [184,190], dabrafenib [41])
FDA-approved in cancer, selumetinib in phase 3
FDA-approved in cancer
NFkB pathwayNFkB-inducing receptor stimulation1. TNFR superfamily stimulation
2. PRR Receptor stimulation
3. IL-1 receptor stimulation
TNFα [40]Phase I-III trials in cancer, all localized infusions
IKK/IkBα1. Boosting IKK-activity
2. Degradation of IkBα
Betulinic acid [45]Phase I trial, Anxiety
IKKBoosting IKK-activityCalcium/calcineurin + prostratin [51]Pre-clinical
Nedd41. Degradation of N4BP1
2. Degradation of TRAF3
p50, RelA, and LMP-2/-7/-101. Enhanced expression of p50
2. Enhanced expression of RelA
3. Enhances expression of LMP-2/-7/-10
Retinoids [41,218]FDA-approved in several diseases, including cancers
Type I IFN pathwayIFN-inducing receptor stimulation1. PRR Receptors stimulation (e.g., RNF135, TRIM25, ISG15, NAB2)
2. IFNA Receptor stimulation
IFNα [100,101]
IFNβ therapy [101]
FDA-approved in hepatitis B & high-risk melanoma
Phase III trial in relapsing multiple sclerosis, Phase I in refractory solid tumors
Type II IFN pathwayIFN-inducing receptor stimulationIFNG Receptor stimulationIFNγ-1b
NK cell therapy [19,33,103,106,107,219]
FDA approved as localized injection in chronic granulomatous disease (CGD) and severe, malignant ostepetrosis (SMO), phase I-III in cancers
Phase I/II Trial in several cancer types
NFkB- and type I IFN pathwaySTING1. Phosphorylation and activation of RelA
2. Phosphorylation and activation of IRF3
STING agonists (e.g., SB 11285 [177])Phase I Trial in patients with advanced solid tumors
NFkB- and type I IFN pathway 1. Increasing APM expression (Calreticulin, TAP2, calnexin)
2. Enhancing type I IFN signaling
3. NFkB stabilization
4. IFNβ secretion
Docetaxel [205]
5-Fluorouracil [206]
Topoisomerase inhibitors (e.g., topotecan [207,209], irinotecan [208], and etoposide [207]
Microtubule stabilizers (e.g., paclitaxel [207,209] and vinblastine [207])
Cisplatin [207]
All FDA-approved in cancer
Epigenetic modificationNFkB and type I/II IFN pathway 1. Histon acetylation to decrease genome accessibility
2. DNA methylation to decrease genome accessibility
HDAC inhibitors: (e.g., Romidepsin, Vorinostat [212] and Panobinostat) [1,211,212,213,214]
DNMTis (e.g., 5-azacytidine [212], Decitabine [54], and Guadecitabine [216])
All FDA-approved in cancer
FDA approved, Guadecitabine phase II trials in cancer).

Share and Cite

MDPI and ACS Style

Cornel, A.M.; Mimpen, I.L.; Nierkens, S. MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers 2020, 12, 1760. https://doi.org/10.3390/cancers12071760

AMA Style

Cornel AM, Mimpen IL, Nierkens S. MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers. 2020; 12(7):1760. https://doi.org/10.3390/cancers12071760

Chicago/Turabian Style

Cornel, Annelisa M., Iris L. Mimpen, and Stefan Nierkens. 2020. "MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy" Cancers 12, no. 7: 1760. https://doi.org/10.3390/cancers12071760

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop