Next Article in Journal
Ca2+-Driven Selectivity of the Effect of the Cardiotonic Steroid Marinobufagenin on Rabbit Sinoatrial Node Function
Next Article in Special Issue
Immunotherapy around the Clock: Impact of Infusion Timing on Stage IV Melanoma Outcomes
Previous Article in Journal
Roles of Extracellular Vesicles on the Progression and Metastasis of Hepatocellular Carcinoma
Previous Article in Special Issue
Fundamental Characterization of Antibody Fusion-Single-Chain TNF Recombinant Proteins Directed against Costimulatory TNF Receptors Expressed by T-Lymphocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insight into Cancer Immunity: MHCs, Immune Cells and Commensal Microbiota

School of Life Science, Guangzhou University, Guangzhou 510006, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work.
Cells 2023, 12(14), 1882; https://doi.org/10.3390/cells12141882
Submission received: 12 May 2023 / Revised: 16 June 2023 / Accepted: 5 July 2023 / Published: 18 July 2023

Abstract

:
Cancer cells circumvent immune surveillance via diverse strategies. In accordance, a large number of complex studies of the immune system focusing on tumor cell recognition have revealed new insights and strategies developed, largely through major histocompatibility complexes (MHCs). As one of them, tumor-specific MHC-II expression (tsMHC-II) can facilitate immune surveillance to detect tumor antigens, and thereby has been used in immunotherapy, including superior cancer prognosis, clinical sensitivity to immune checkpoint inhibition (ICI) therapy and tumor-bearing rejection in mice. NK cells play a unique role in enhancing innate immune responses, accounting for part of the response including immunosurveillance and immunoregulation. NK cells are also capable of initiating the response of the adaptive immune system to cancer immunotherapy independent of cytotoxic T cells, clearly demonstrating a link between NK cell function and the efficacy of cancer immunotherapies. Eosinophils were shown to feature pleiotropic activities against a variety of solid tumor types, including direct interactions with tumor cells, and accessorily affect immunotherapeutic response through intricating cross-talk with lymphocytes. Additionally, microbial sequencing and reconstitution revealed that commensal microbiota might be involved in the modulation of cancer progression, including positive and negative regulatory bacteria. They may play functional roles in not only mucosal modulation, but also systemic immune responses. Here, we present a panorama of the cancer immune network mediated by MHCI/II molecules, immune cells and commensal microbiota and a discussion of prospective relevant intervening mechanisms involved in cancer immunotherapies.

1. Introduction

In recent decades, cancer immunotherapies like chimeric antigen receptor-engineered T cell (CAR-T), checkpoint blockade of PD-1/PD-L1, etc., are emerging in an endless stream which arrests the attention of physicians, patients and researchers from all over the world for their different, genius and extraordinary efficiency on cancer treatment compared with traditional chemotherapy and radiotherapy. This demonstrates that cancer generation and development are closely bound up with immunological events in the body.
In general, extrinsic pathogens and abnormal intrinsic cells with senescent, malignant transforming, infected, dysfunctional or other phenotypes should be targeted by immune cells and, in consequence, be killed. In specific immune responses to the special antigens, there are major histocompatibility complexes, commonly including MHC-I and MHC-II molecules that play key roles in cell-intrinsic and pathogen-extrinsic antigen targeting and presentation, respectively. All nucleated cells in the body express MHC-I molecule and cellular-intrinsic antigens are thereby processed and presented by MHC-I. For example, an infected cell combines pathogen peptide with MHC-I and presents the complex, or a malignant cell combines cancerous peptide with MHC-I and presents the complex. Those complexes can be recognized by cytotoxic T lymphocytes (CTLs), such as CD8+T cells, and apoptosis of the targeted cell is initiated as a consequence, while extrinsic pathogens, including bacteria, fungi, viruses, chlamydia or mycoplasma, can be recognized and engulfed by antigen-presenting cells (APCs) via endocytosis. Thereafter, extrinsic peptides are presented by APCs through being combined with MHC-II molecules. Usually, MHC-II molecules are identified by CD4+T cells which then play a role as a helper in mediating CD8+T cell activation and inducing B lymphocyte differentiation, and then the latter two initiate cellular immunity and humoral immunity, respectively.

2. MHC-I Reduction and Immune Evasion

In cancer immune surveillance, T lymphocyte-mediated specific immune response plays a crucial role. However, cancer immunity can be blocked through the alteration of the tumor microenvironment, leading to surveillance evasion. Thus far, the related strategies include: decreasing cancer antigens’ immunogenicity and rendering cancer cells less visible, muting APCs, inhibiting naïve T cell stimulation, depressing CTL infiltration into the tumor, suppressing CTLs’ cytotoxic efficacy, etc. [1,2,3]. Clinical cancer immunotherapies may, therefore, suffer from cancer cell-mediated immune invisibility through immune evasion mechanisms, which usually result from the downregulation of MHC-I expression or the failure of antigen presentation (Figure 1). In human cells, MHC-I molecules are known as human leukocyte antigens (HLA) and HLA class-I molecules are composed of classical (class-Ia) and the non-classical (class-Ib) components [4,5,6,7]. In fact, HLA-Ia expression defects are always found followed by uncontrolled growth of both primary and metastatic cancers [8,9,10], demonstrating their correlation with high tumor grading and progression, decreased survival and ineffective immunotherapies [11,12,13,14]. An MHC-I expression defect could derive from reversible lesions and irreversible lesions. The former are aroused from epigenetic modifications such as IFNγ-induced MHC-I expression [15,16], while the latter are aroused from less common structural mutations. Through investigating the regulation of MHC-I expression, Kobayashi et al. found that NOD-, LRR- and CARD-containing 5 (NLRC5) notably upregulated MHC-I, β2-microglobulin (β2M) and transporter associated with antigen processing 1 (TAP1), while the latter two as indispensable parts play key roles in the MHC-I antigen presentation pathway [17]. This study also revealed that NLRC5 was induced by IFNγ in a dose-dependent manner, indicating that NLRC5 probably plays a key role in the MHC-I antigen presentation pathway. Subsequently, they also reported in the following studies that NLRC5 expression was lost in varying cancers and the loss was correlated with the loss of CTL activation in several types of cancer [18].
It seems that NLRC5 expression is not invariable in different tissues according to the studies: high levels were shown in almost hematopoietic cells and tissues, particularly in CD8+ and CD4+ T lymphocytes, B lymphocytes, Natural Killer (NK) cells and NK-T [19,20,21,22,23], whereas intermediate levels were shown in macrophages and dendritic cells [21,24,25,26]. Upregulated MHC-I expression is performed not only through the NLRC5 pathway itself, but also in a synergistic way with the help of additional cis-regulatory elements’ (ISRE and enhancer A) activation. Their expression is induced by IFNγ-caused STAT1 homodimer formation, which activates IRF1 and NLRC5 gene promoters through binding gamma-activated sequence (GAS) [27]. This was also confirmed by the involvement of activated IFN and STAT1 in the increase in NLRC5 expression during the activation of CTLs and T helper (Th) cells [21]. In vitro, the efficiency of TCR transgenic CD8+ T cells was activated and supported by NLRC5-deficient bone marrow-derived macrophages (BMDM) provided with SINFEKKL peptide, which was as similar as the efficacy of CD8+ T cells induced by wild-type BMDCs, while it was failed with the application of NLRC5-deficient cells provided with negative control [28]. After in vivo intravenous loading of Listeria monocytogenes, the IFN-producing CD8+ T cell was not aroused to an increase in the cell number in the spleen or the liver of NLRC5-deficient mice, consequently resulting in severe infection compared with the CD8+ T cell in wild mice [28,29]. In addition to CD8+ and CD4+ T lymphocytes, NLRC5 is implicated in γδT cell activation as well and its expression positively correlated with the induction of butyrophilin (BTN) family protein BTN3A1-3, whose gene contains SXY modules W/S, X1, X2 and Y boxes as cis-regulatory elements of the MHC-II gene promoter [30]. All these studies suggest that NLRC5 plays a role in the activation of MHC-I transcription, and the facilitation of CD8+ and CD4+ T lymphocyte development and immune function initiation (killing or helper).
In spite of numbers of studies proving NLRC5 expression in different tissues and its role in the MHC-I antigen presentation pathway [19,20,21,22,23], the regulatory mechanisms of the expression have not been well-elicited. These studies suggest that Chromatin NLRC 5 promoter accessibility is varied and modulated by different transcription factors in different cell types, where epigenetic regulation of chromatin remodeling may be a key pathway potentially adjusting NLRC 5 expression, such as in human cancers as well as cancer cell lines. NLRC 5 was involved in MHC-I expression attenuation through regulating promoter hypermethylation [18]. NLRC 5 expression, like other functional proteins, can also be regulated at the post-transcriptional and post-translational levels by long non-coding RNAs (lncRNAs) [31,32,33,34,35,36]. Interestingly, it was recently reported that MHC-I and APM expression was reduced by depressing NLRC5 in quiescent hair follicle stem cells and muscle stem cells in order to protect from immune surveillance and destruction [37]. Additionally, the reduction of MHC-I in cancer-originating cells of different types of cancer disrupted CTL-mediated damage, leading to the formation of variants that could escape immune surveillance and thereafter causing cancer recurrence [38,39,40,41,42,43]. Compared with immune evasion, these may suggest that NLRC5 and MHC-I reduction may facilitate the formation of a global dedifferentiation phenotype in cancer cells, which suggests the progression of cancer growth and the initiation of cancer cell communities.
As NLRC5 plays key role in the activation of MHC-I expression which is commonly lost in cancer immune evasion, NLRC5 could be the point that can switch on antitumor immunity. Several relevant in vitro and in vivo studies suggested that MHC-I expression in tumor cells could be more necessary than that expression in APCs to induce protective antitumor immunity [44,45]. Therefore, NLRC5 expression acquisition in tumor cells can enhance their MHC-I expression and antigen presentation and put forward antitumor immune response and CTL-mediated destruction [46,47,48]. However, in contradiction with the studies above focusing on the antitumor aspects of NLRC5, there are also studies implicating the role of NLRC5 in promoting tumor growth. According to their reports, NLRC5 was shown to participate in vitro in modulating the biological behaviors of HCC, ccRCC, glioma and ESCC cells, such as promoting cancer cell growth, motility and migration, and played a similar role in mice lacking the functionality of an adaptive immune system [33,36,49,50,51,52,53], while NLRC5 in mice with a competent immune system evidently suppressed the growth of melanoma and pancreatic ductal adenocarcinoma (PDAC) cells [54,55]. Considering that NLRC5 and MHC-I could be involved in progressive cancer growth and cancer cell population initiation, those contrary phenomena may result from that NLRC5-elicited antitumor immune response overcoming NLRC5-stimulated cancer growth in cancer cells originating from mice with normal immune function.
Studies on how to compensate for insufficient MHC-I expression and blocked antigen presentation in tumor cells are still progressing. IFN has been shown to potentially reverse the defect in MHC-I expression and accelerate antigen processing in cancer cells through the modification of proteasome constituents [45]. However, the limitation of potential use of IFN is there, where its systemic toxicity and development of cancer unresponsiveness to IFN are involved [56,57,58]. Epigenetic drugs may derepress NLRC5 showing the capacity to restore MHC-I expression, while their efficacy on solid tumors is limited and off-target effect is also a key challenge [59,60,61]. In general, genetic and epigenetic alteration-caused loss of NLRC5 paves the way for MHC-I deficiency, which is a common mechanism of cancer unresponsiveness to immunotherapy.

3. TsMHC-II Function and Tumor Immune Surveillance

Whereas MHC-Is are distributed primarily on the surface of almost all nucleated cells and mainly provide endogenous antigen to CD8+ T cells, MHC-IIs of APCs, such as dendritic cells (DCs), B cells and macrophages, primarily present exogenous antigens to CD4+ T cells. Though MHC-I-restricted neoantigens have shown to be important effectors for tumor-specific CTLs in the success of immune checkpoint inhibition (ICI) [62,63,64,65,66,67,68,69], prognostic indicators are not precise enough [70,71,72,73,74]. Therefore, considering CD4+ T cells play important auxiliary roles in the activation of CD8+ T cells and the formation of memory T cells [75,76,77,78,79,80], the influence of MHC-II-restricted CD4+ T cells has been addressed and recognized as being effective [81,82]. In fact, besides APCs, other cell types including tumor cells are able to express MHC-II as well [71]. Tumor-specific MHC-II expression (tsMHC-II) can facilitate immune surveillance to detect tumor antigens (Figure 2), and thereby has been used in immunotherapy, including superior cancer prognosis, clinical sensitivity to ICI therapy and tumor-bearing rejection in mice [72,73,74,75,76,77,81,82]. Clinically, tsMHC-II has been seen in a variety of human tumors including: breast cancer [73,74,83,84,85,86,87,88,89,90], colorectal cancer [91,92], classic Hodgkin lymphoma [76], glioma [93], melanoma [72,77,94], non-small cell lung cancer [95], ovarian cancer [96,97] and prostate cancer [98].
In the mechanism, those functions of tsMHC-II may be derived from the stimulation of CD4+ T cell subsets, of which Th1 CD4+ cells express IFN-γ and other cytokines, while regulatory T cells (Tregs) play a suppressive role in immunity and inflammation and contribute to tolerance [99]. For other CD4+ T cell subsets like Th2 and Th17, the mechanisms are still ambiguous for their both pro-tumor and anti-tumor efficacy in cancer immunity [99]. Usually, antigens can be presented by MHC-II from a greater repertoire. This causes CD4+ T cells to be capable of recognizing cancer-associated antigens in a wider array than CD8+ T cells [100,101,102]. As tumors have fewer candidate neoantigens compared with normal cells [80], this specially indicates the functional significance of CD4+ T cells for tumor treatment. However, the exact effect of tsMHC-II on CD4+ Tregs in tumors is still not clear. In Tregs, TCR stimulation is performed through MHC-II as well to maintain suppressive activity [103], while in tumors in view of Tregs abundance [104,105] and tsMHC-II effect of improving immune-mediated outcomes, it could be suggested that tsMHC-II may be unable to stimulate TCR in Tregs. Normally, signal 1 (MHC:T cell receptor binding) and signal 2 (co-stimulation with CD80/86 binding to CD28) are required for T cell activation [106], while CD80/86 is not typically expressed by tumor cells [107]. Instead, non-classical co-stimulatory receptors beyond CD80/86 can be expressed by some cancer cells and are sufficient to activate certain T cell subsets [71,107], for example CD70, OX40-ligand, as well as molecules from adjacent, juxtaposed immune cells in the tumor microenvironment [108,109,110].
The expression of MHC-II as well as antigen presentation machinery is triggered by the transcriptional master regulator Class II Transactivator (CIITA). Without directly binding DNA, CIITA recruits transcriptional factors at promoter regions of MHC-II-related loci through scaffolding activity and induces the antigen presentation pathway where CIITA is necessary and sufficient [75,111]. CIITA plays a particularly important role in the process of IFNγ-initiated MHC-II expression, while some cancer cells appear insensitive to IFNγ stimulation [72,112]. Instead, multiple other pathways may be involved in regulating MHC-II expression in those cells. For example, JAK/STAT signaling is necessary for the upregulation of MHC-II expression [113,114], and MHC-II was suppressed by RAS/MAPK activation in breast cancer [83]. MAPK stimulated the CIITA promoter pIII in the HLA-DR+ melanoma cell line, and this promoter initiates transcription primarily in B cells [115]. Meanwhile, tumor cells may also downregulate MHC-II expression through some mechanisms. In Hodgkin Lymphoma, CIITA expression alteration was suffered in genomes, while tsMHC-II expression was blocked in primary large B-cell lymphoma [116,117,118]. Other non-genomic mechanisms are also involved. L-myc, N-myc and human achaete-scute complex homologue-1 were overexpressed and then interrupted the transcriptional response to IFNγ through binding to promoter IV of CIITA in small-cell lung cancer and neuroblastoma [95,119]. In addition, epigenetic alteration is a potential mechanism as well being involved in the reduction in MHC-II expression in human tumors [120]. This could link to cancer immune evasion since tumor cells can avoid recognition by anti-tumor CD4+ T cell subsets through inhibiting MHC-II expression. Therefore, repression of tsMHC-II expression may result in the resistance to ICI, and ensuring the correlation of the level of tsMHC-II expression with sensitivity to immunotherapy should be key to treatment and prognosis.
In fact, there could be a consequent correlation between tsMHC-II upregulation and tumor immunogenicity increase, suggested via the studies in mice. In clinic, tsMHC-II usually indicates improved prognosis, upregulation of favorable response to ICI, enlargement of TILs and pro-inflammatory IFN signaling in human tumors. This renders tsMHC-II a clinically operational biomarker reflecting sensitivity to the ICI, and strategies designed to increase the expression of the tsMHC-II could increase sensitivity to immunotherapy. Moreover, studies via murine models also infer that tsMHC-II should be a potential tool for anti-tumor vaccines. Mice were protected from injection with parental tumor cells through previous inoculation with non-viable tumor cells that express MHC-II [121,122,123,124]. Since studies showed that tsMHC-II upregulation often brings about tumor rejection, we may have a hypothesis that tsMHC-II expression promotion in human cancers could facilitate anti-cancer immunity and enhance sensitivity to ICI. Developing strategies to upregulate the levels of tsMHC-II in tumors through treatment, and gaining insights into its biological function, may bring new insights in cancer immunotherapy, but this requires more rigorous testing.

4. NK Cells and Anti-Cancer Immunity

While the priming of MHC-I expression and neoantigen presentation is known to introduce tumor antigen recognition performed by CTLs and subsequently trigger potent antitumor response, there are a number of strong scientific findings showing that immune responses elicited by non-CTLs like Natural Killer (NK) cells also play key roles in the regulation of cancer immunity. NK cells are a small subset of lymphocytes that account for innate immune response, in part including immunosurveillance and immunoregulation. NK cells are the early and rapid responders of the innate immune system and are inherently equipped with the capacity to recognize and capture pathogen-infected, damaged or transformed cells independent of the prophase sensitization process or antigen presentation. This distinct surveillance of NK cells is performed through the signaling that is triggered from surface-activating receptors or inhibitory receptors and most of the receptors can recognize molecules in a large repertoire including MHC-I, HLAs, chain-related proteins A and B molecules, nectin family proteins and others. Usually, the cytotoxic response of NK cell depends on the balance between the activating and inhibitory signals they receive (Figure 3). Responses are triggered under the condition of excessive activating signals [125]. NK cells also express FcgRIII receptors, which can recognize antibodies that are specific to tumor antigens and thereby perform antibody-dependent cell-mediated cytotoxicity (ADCC).
Compared with T cells that are targeting in an antigen-specific manner, NK cells, without targeting unique antigens, recognize expression patterns indicative of malignant transformation, that is to say, the pattern of the activating signals, such as the abnormal expression pattern of the molecules on the surface of a tumor cell could be preferentially recognized compared with the normal pattern of a healthy cell. The board of NK cells’ recognition is not specific, but broad, without prior training and independent on one unique molecule. Cytotoxic response triggered by NK cells could not be attenuated in case of the deficiency of unique antigens. Therefore, with similar functions and effects as CTLs, NK cells are capable of eliciting immune response to drug-resistant cancer cell populations. In contrast to CD8+ T cells, MHC deficiency annuls inhibiting signals towards tumor cells in NK cells and renders them more activating signals towards tumors. With this capability, NK cells have been shown to have exclusive capabilities to target highly malignant cancer stem-like cells and tumors with poorly differentiated or undifferentiated histological stages that are highly resistant to chemotherapy. Secondly, NK cells can catalyze differentiation of tumor cells through IFNγ secretion and stimulation [126], which causes remodeling of the surface receptor pattern of tumor cells, including an upregulation in MHC-I and CD54 and a downregulation in CD44 expression, followed by the disturbance of tumor growth and metastasis [127] and being well-recognized and eliminated by T cells. Thirdly, NK cells can selectively target aging tumor cells. Ruscetti et al. observed that KRAS-mutant lung tumor growth in mice was reduced after treatment with a cytostatic drug regimen with the effect that could primarily lead to natural aging of the body [128]. The evidence above supports the involvement of NK cells in checkpoint blockade and their impact on the response to immunotherapy.
In the immune system, NK cells participate in the initiation of the adaptive immune response by recruiting other immune cells, like DCs. In fact, through NK–DC crosstalk, NK cells release IFNγ and TNFα that promote DCs’ maturation [129], while DCs can facilitate NK cell proliferation, survival and activation through THE secretion of cytokines such as IL-12 and IL-15 [129,130]. Moreover, DCs, in case of misleading to maturation, will be killed in DC editing, which is performed by NK cells through the engagement of the activating receptor NKp30 [131]. Therefore, this crosstalk is of crucial significance in activating innate immune responses along with immune response to tumors. In the latter, DC–NK crosstalk has been considered an indicator of cancer immunotherapy after years of investigation [132,133]. Conventional type 1 DCs (cDC1) are important players in regulating cross-priming activation of T cells located in tumor-draining lymph nodes through secreted chemokines and cytokines that are involved in the modulation of T cell viability, capability and infiltration into the tumor microenvironment, which presents the importance of their clinical application for cancer immunity through highlighting their effect on tumor rejection abolishment, responsiveness to CAR-T therapy and ICI in mice with cDC1 deficiency [134,135]. For example, a recent study reported that induction and activation of cDC1s that reside within the tumors overcame acquired resistance and facilitated the tumors’ responsiveness to anti-PD-L1 therapy [136]. Actually, after activation, NK cells produce cDC1 chemoattractants and secrete them into the tumor microenvironment to attract cDC1s, and thereafter cDC1s can recruit CTLs and, in turn, elicit an adaptive immune response [137]. In clinic, increased populations of both tumor-infiltrating NK cells and cDC1 cells in the tumor microenvironment have been used as a biomarker for predicting and evaluating tumor responsiveness to PD-1 immunotherapeutic agents and prognosis of melanoma [138].
Oncolytic viruses (OVs), as a novel sort of anti-cancer drug, were first approved in 2015 and were followed rapidly by numerous other clinical trials. Originally, the therapeutic effect of Ovs was regarded to be due to viral completing replication within tumor cells, which in turn led to cell lysis. Now, it is recognized that OVs largely contributing to the induction of adaptive and innate immune responses could be responsible for the OV agents’ efficacy that has been reported [139]. That is to say, OVs can reduce the immunosuppressive pressure in the tumor microenvironment by priming the immune system. In clinic, NK cells are potentially involved in Ovs’ therapeutic efficacy via antiviral response of the innate immune system. Usually, the dual role of NK cells in innate defense leads to the removal of malignant cells and virus-infected cells, which meanwhile results in the clearance of the virus through killing virus-infected cancer cells and, thus, limits Ovs’ efficacy. Therefore, promoting OV-mediated lysis of tumor cells through endogenous NK cell depletion, together with adjuvant exogenous NK cell injection that augments the capacity of the immune system, could improve the efficacy of immunotherapy [140]. This was confirmed in a primary glioma mouse that was granted a significantly longer survival after being treated with the combination of OV therapeutics with activated human NK cells [141]. A better understanding and arrangement of the therapeutic variables such as Ovs’ nature, tumor stage, histological grade, dose regimen and adverse effects of the adoptive NK cell therapy could be necessary to optimize therapeutic interventions and improve the efficacy of antitumor immunity.
The efficacy of cancer immunotherapy has been clearly demonstrated to associate with the function of NK cells. Moreover, the incidence of cancers or other diseases could also be increased in case of NK cell dysfunction. In a prospective cohort study composed of healthy individuals, the capacity of immune response and disease incidence were investigated, showing that participants with low CTL cytotoxic activity had a higher risk for cancer incidence compared with those with medium or high cytotoxic activity [142]. Clinically, NK cells are dysfunctional or occur less frequently in most cancer patients and are further affected by surgical treatment and standard procedure of chemotherapy [143,144]. For example, fructose-bisphosphatase 1 could be inhibited by glycolysis, which leads to the dysfunction of NK cells and further tumor progression in KRAS-driven models of lung cancer [145]. Moreover, altered patterns of protein expression on the surface of tumor cells could also be the cause of dysfunctional NK cell response, such as radiation-caused increased PD-L1 level and decreased ligand level for Natural Killer Group 2D (NKG2D) receptor-protected non-small cell lung cancer cells from NK cell cytotoxicity [145,146,147,148]. Thus, surgery or treatment-caused NK cell dysfunction could pave the way to limited response or compromised efficacy to immunotherapies or related therapies targeting immune cells or molecules. In the adoptive NK cell therapy, endogenous NK cells were provided with a critical barrier in the setting of an altered tumor environment to attenuate their sensitivity to tumor immunosuppression. In addition, these cells were highly cytotoxic and targeting was ameliorated by chimeric antigen receptors to effectively increase the rates and duration of response [149,150].

5. γδ. T Cells and Anti-Cancer Immunity

According to the surface expression of αβ and γδ TCRs, T cells are primarily classified into two subsets: αβ T cells and γδ T cells. Whereas αβ T cells elicit an adaptive immune response through the recognition of target antigens that are presented via MHC molecules, γδ T cells account for 1–5% of total T cells compared with them, and are involved in the innate immune response that functions in a manner not by the MHC-introduced antigen presentation pathway but through the recognition of the surface proteins of the targeted cells, such as BTN and CD1 molecules (Figure 4) [151]. In addition, γδ T cells can bring us a new opportunity for “off-the-shelf” cellular therapeutics in allogeneic adoptive transfer settings since there is a low risk of the incidence of graft-versus-host disease with the application of γδ T cells clinically [152,153]. Currently, αβ T cells have been the focus of the majority of studies on T cell immunology and clinical application, whereas γδ T cells have also shown their obvious involvement in cancer immune processes and their potent killing effects on various types of cancer as reported by past studies [154,155,156,157]. For example, by analyzing the correlation between 22 different leukocyte subsets and cancer survival in 5782 clinical tumor samples, Gentles et al. proposed that tumor-residing γδ T cells might be the best predictor of overall patient survival [158].
The definition of human γδ T cell subtypes usually depends on the δ chain of γδ TCR, and the most relevant isoforms are, accordingly, Vδ1-3, where the most comprehensively reported isoforms are Vδ1 T cells and Vδ2 T cells, which both arise during the period of fetal development [159]. In adults, Vδ1 T cells maintain a population of about 10–15% of γδ T cells, primarily residing in peripheral tissues such as the gut epithelia, dermis, spleen and liver [160,161]. Vδ2 cells make up 80–85% of the total peripheral γδ T cells and are progressively predominant in the blood and spleen in adulthood [160]. The remaining γδ T population comprises primarily Vδ3 T cells whose lineage and rearrangement are still less well-known [162]. During developmental stages, TCR Vδ2 strands often connect to Vγ9 chains to form Vγ9Vδ2 T cells, which constitute the major component of the γδ T cell population in human peripheral blood [163]. As usual, Vγ9Vδ2 T cells have the ability to recognize infected or transformed cells enriched for phosphorylated antigens (pAgs) or intermediate metabolites such as isopentenyl pyrophosphate (IPP). Tumoral pAgs bind the intracellular domain of BTN3A1, which is associated and comprises complexes with BTN2A1, and BTN2A1 can be recognized directly by TCR Vγ9 chain [164], while IPP is overproduced by the dysregulated mevalonate pathway in cancer cells, which activates Vγ9Vδ2 T cells [165,166]. Instead of pAg-mediated BTN recognition by Vγ9Vδ2 T cells, non-Vγ9Vδ2 T cells with more diverse TCRs usually recognize other molecules like CD1, a family of surface glycoproteins that present with lipid antigens [167]. For example, it has been shown that Vδ1 and Vδ3 subsets recognize CD1 proteins [167,168,169,170,171]. Additionally, BTN molecules were found to be produced within hematological and solid tumors, while CD1 mainly serves as a molecular marker for tumor cells derived from the myelomonocytic and B-cell lineage and is rarely expressed within solid tumors [172,173]. In addition to TCRs, other surface molecules are also expressed by γδ T cells with the purpose of providing additional stimulatory signals and play a role in tumor cells’ recognition as well. Those molecules are usually Natural Killer receptors, such as NKG2D, DNAX Accessory Molecule-1 (DNAM-1), NKp30, NKp44 and NKp46. The ligands for these receptors are often overexpressed by malignantly transformed cells, which are thereby targeted by them and activate them accordingly [174,175,176,177,178,179].
In general, γδ-T cells can use multiple pathways to exert anti-tumor cytotoxic effects. First, activation of the receptor induces perforin expression and release of granzyme B, thereby mediating pore formation at the tumor cell membrane and leading to the entry of granzyme protease into the cell, which subsequently initiates apoptosis of tumor cells [180]. Secondly, TNF-related apoptosis-inducing ligand (TRAIL) and Fas ligand (FasL) expressed by γδ T cells recognize and bind their respective receptors expressed on tumor cells, thus resulting in the induction of apoptosis [180,181,182]. Thirdly, tumors can be targeted by γδ T cells via ADCC. For example, CD16 (Fc receptor III) is expressed on γδ T cells and binds the Fc region of antibodies that is bound to the relevant antigen of the tumor cell, subsequently leading to tumor lysis [180,183,184]. Another co-stimulator is a Toll-like receptor (TLR) that can activate γδ T cells either directly or by participating in the activation of DCs [185]. TLRs feature their structure with integral membrane glycoproteins and thereby are capable of recognizing prototype patterns [185,186]. In response to stimulants such as specific lipid, peptide or nucleic acid PAMPs (pathogen-associated molecular patterns) and DAMPs (danger-associated molecular patterns from damaged tissues), TLRs dimerize and then trigger subsequent signal transduction pathways that lead to the upregulation of inflammatory factors’ expression, like TNF-α, IL-6, IL-1β, IL-12 and IFN-γ [185,186]. However, the consequence of TLR signaling depends on the specific TLR and surrounding circumstances, which can lead to the activation of the immune system or else the suppression.
Cancer cells constitute the main components of the tumor microenvironment, which is surrounded by stromal cells or tissues such as fibroblasts and blood vessels. The microenvironment is usually infiltrated by immune cells and characterized by hypoxia and immunosuppression [187]. Compared with other immune cells that can be hindered by those unfavorable conditions, γδ T cells seem to be more adaptable to this harsh growth environment with hypoxia and inhibition, and still can efficiently identify relevant markers expressed in tumor cells, complete MHC-independent activation and express inflammatory cytokines [188]. It was also shown that activated γδ T cells can stimulate other immune cells and coordinate an antitumor response with them accordingly. For example, M1 and M2 macrophages that infiltrate into the tumor microenvironment could be targeted by γδ T cells and encounter microenvironmental-specific chemokines. Those molecules are able to polarize macrophages towards a pro-tumor M2 state and repolarize them to an anti-tumor M1 state [189]. Additionally, γδ T cells have sorts of immunomodulatory interplay with other immune cells. Vγ9Vδ2 T cells act as APCs, functioning after being activated in a manner similar to DCs, including the increased expression of MHC-I and MHC-II molecules and the release of APC-associated adhesive and costimulatory molecules [190,191]. They may promote their capacity of antigen presentation from both apoptotic and live cancer cells through expressing scavenger receptor CD36 [192]. Then, similar to DCs, the antigen presentation accordingly triggers the differentiation and proliferation of naive CD4+ and CD8+ αβ T cells [190,191]. Another well-characterized interaction is the crosstalk between γδ T cells and DCs [193], during which Vγ9Vδ2 T cells provide assistance to DC maturation through TNF-α and IFN-γ production, rendering co-stimulatory molecule expression increased in DCs, cytokine secretion enhanced and, as a consequence αβ T cell proliferation and IFNγ production, induced effectively [194]. Through the crosstalk, DCs after maturation activate γδ T cells in turn. γδ T cells also interact with NK cells for their activation. Maniar et al. showed that NK cell cytotoxicity against NK-resistant tumors was increased after NK cells co-cultured with IPP-expanded Vγ9Vδ2 T cells [195]. The crosstalk was performed through co-stimulatory interaction between CD137L on γδ T cells and CD137 on NK cells, which facilitated NK cell activation and the subsequent augmentation of NKG2D expression and increase in antitumor cytotoxicity. An in vivo study as further evidence was reported, showing a key role for γδ T cells in inducing IFNγ production in NK cells during early immune responses [196].

6. Eosinophils and Tumor Microenvironment

Eosinophils are a special class of granulocytes that are differentiated in response to allergic reactions, abnormal skin conditions, parasitic and fungal infections, autoimmune diseases, certain cancers and bone marrow diseases. Eosinophils are generally produced in the bone marrow through the differentiation from distinct CD34 myeloid progenitor cells, and normally reside in the connective tissues, specifically in the thymus, gastrointestinal tract, spleen, lymph nodes, ovaries and uterus [197]. Since the late 1900s when eosinophilia was identified in the peripheral blood of cancer patients [198], the tumor-infiltrating eosinophil microenvironment has been shown in sorts of solid tumors, particularly in the tumors of mucosal organs like the gastrointestinal tract and the lungs [199], suggesting that eosinophils are involved in cancer-inflammatory response or are recruited in response to therapies [200].
Clinical and experimental results provided us the potential involvement of eosinophils in the interactions between eosinophils and lymphocyte subsets, including T cells, NK cells and innate lymphoid cells (ILCs) (Figure 5). ILC2s play a role in the mediation of type 2 immune responses and induce chemotactic eotaxin production in epithelial cells and macrophages [201,202] that triggers eosinophil expansion or migration, thereby contributing to the tumor microenvironment in an eosinophil-dependent way or in an eosinophil-independent fashion [203]. The eosinophil–ILC2 interactions were studied mostly in experimental melanoma [204]. For example, one study found that intravenous injection of B16-F10 melanoma cells into mice led to an increase in eosinophilic infiltration into the lungs and upregulated production of IL-5 in lung ILCs [205]. Moreover, in mice lacking IL-5 or treated with anti-IL-5 antibody, the intravenous injection of B16-F10 cells resulted in the decrease in eosinophil infiltration and increase in lung tumor colonization compared with wild-type mice [205]. This suggests that the increased eosinophilic infiltration into the lungs was associated with IL-5 production. In addition, GM-CSF and IL-33 were also shown to be involved in the regulation of ILC2–eosinophil cross-talk in the tumor microenvironment [206]. GM-CSF-deficiency increased tumor growth in tumor-loading mice, and the contribution of GM-CSF production by ILC2s was concerned with the increase in eosinophils and the decrease in tumor growth in tumor-loading GM-CSF-deficient mice [204]. In a lung metastatic murine model by intravenous injections of B16-F10 melanoma cells or Lewis lung carcinoma, ILC2s, eosinophils, CD8+ T cells and NK cells were activated and tumor-infiltrating by IL-33 overexpression or IL-33 treatment [207,208].
Eosinophils can interact with NK cells as well through expressing multiple receptors and ligands, such as a CD2 subfamily of receptors, LFA-1. The receptors are involved in the mediation of eosinophil–NK cell cross-talk, while the interruption of eosinophil ligands activating natural cytotoxicity receptors NKp30 and NKp46 in vitro inhibited eosinophil-mediated NK cell activation [209,210,211]. Intravenous injection of 4T1 tumor cells rendered NK cells recruited to the lungs through IL-33 stimulation, which depended on C-C motif chemokine ligand 5 (CCL5) expressed by eosinophils and CD8+ T cells. NK cell recruitment was reversed after deleting eosinophils and CD8+ T cells [212]. A tumor-cytotoxic effect against K562 myelogenous leukemia cells was shown after co-culturing IL-12-stimulated NK cells with eosinophils, and this cytotoxicity depended on eosinophils’ contact [211]. However, eosinophils may play a suppressive role in NK cell cytotoxicity under the preexistence of type 2 immune responses, which promote tumor growth as a consequence. ILC2-derived IL-5 induced an eosinophil-mediated suppressive effect on lung NK cell activity through modulating the metabolic environment, while the suppressive effect on IFN-γ and granzyme B production by NK cells was attenuated via anti-IL-5 treatment [213].
Bioinformatics data indicated that eosinophils may interact with T cells in cancer. In various primary tumors, through the prediction of the corresponding RNA signatures of multiple immune cells by bulk RNA sequencing and an RNA profiling algorithm, eosinophils, the abundance of such immune cells correlated with the abundance of resting memory CD4+ T cells in the primary tumor [199]. Moreover, eosinophil RNA signature may also have correlation with the RNA signatures of CD8+ T cells according to bulk RNA-sequence data from pleural metastatic samples of breast cancer patients [214]. A transcriptional profile characterized by M1 macrophages was acquired in IFN-γ-stimulated eosinophils, which presented increased cytotoxicity against colorectal cancer [215,216]. Chemokine expression, like CCL5, C-X-C motif ligand 9 (CXCL9), CXCL10 and CXCL16, were augmented in eosinophils after IFN-γ stimulation, and those chemokines were capable of recruiting T cells [214]. Accordingly, the adoptive transfer of IFN-γ-activated eosinophils into the lungs of tumor-loading mice promoted the infiltration of CD4+ and CD8+ T cells [214], while pharmacological depletion of regulatory T cells in a tumor model induced evident eosinophilia, which facilitated antitumor immunity [217]. This could demonstrate an anti-tumorigenic function of eosinophil–T cell interactions in the tumor microenvironment. In addition, eosinophils are capable of mediating antigen presentation and, thus, facilitate T cell responses [218].
Clinical studies from independent centers reported that peripheral eosinophil number is upregulated after ICI, especially in the cancer therapies with anti-CTLA4 and anti-PD-1 antibodies [219,220,221,222]. Eosinophils were thereby suggested as cellular biomarkers and end-stage effector cells for positive prognosis in several tumors [223,224,225,226,227,228]. Moreover, human pluripotent stem cell-derived eosinophils presented synergistic tumor-killing capacity with CAR T cells in solid tumors. In the studies with the established tumor-bearing mice loaded with HCT116 (human colorectal carcinoma), MDA-MB-231 (human breast adenocarcinoma) or HepG2 (human hepato-cellular carcinoma), combined therapies with CAR T cells and human embryonic stem cell-derived eosinophils demonstrated superior anticancer efficacy on cancer therapy compared with the single treatment of those alone [229].

7. Microbiota and Anti-Cancer Immunity

The parts of the human body, including the digestive system, respiratory system, immune system and female reproductive system, are more or less characterized with colonization by sorts of microorganisms in their mucosal environment. Most studies focusing on the microbiota of the gastrointestinal system indicate that the gut microbiome is actively involved in several host functions, including nutrient absorption, metabolism, immunity and cancer immunology [230].
In the gut microbiota, bacteria play a role in multiple inflammatory diseases of the gastrointestinal tract, and also in the tumorigenesis of the digestive tract. For example, a pronounced correlation has been suggested between Helicobacter pylori infection and the development of atrophic gastritis, metaplasia, dysplasia and gastric cancer progression [231,232]. Fusobacterium nucleatum has also been shown to be correlated with tumorigenesis in the gut, extensively in colorectal cancer (CRC). An increased abundance of F nucleatum was found in CRC tumors compared with normal tissue. Moreover, enrichment of this species in tumor tissue was involved in the advanced progression from adenomas to carcinomas [233].
In addition, the gut microbiota is also capable of functioning in distance on tumors that are developed in tissues distant from the digestive tract. It has been suggested that Gammaproteobacteria can move from the gut to pancreatic tumors, possibly through the duodenum and pancreatic duct, and was involved in efficacy attenuation of the drug gemcitabine there via metabolizing its active form [234]. Tumor growth was promoted by the treatment with commensal microbiota in a Kras transgenic mouse model of PDAC [235]. In this model, on the one hand, the expression of Kras in the mouse pancreas accelerated the malignant development of pancreatic tumors, and on the other hand, caused species changes in the digestive tract microbiome as compared with littermates of wild-type mice, indicating that the gut microbiome alteration could likely be relevant to the enrichment in tumor-promoting bacteria. In a clinical PDAC study, it was shown that the microbiome species composition in the tumor microenvironment was significantly different from the adjacent healthy pancreatic tissue through 16S rDNA sequencing; in addition, some of the bacteria detected in the tumor microenvironment were also detected in the patient’s feces. Specifically, the flora density of Sachharopolyspora, Pseudoxanthomonas, Streptomyces and Bacillus clausii in the tumor microenvironment was strongly associated with the prolonged survival of patients [236]. Through further in vivo experiments using fecal microbial transplantation (FMT), mice that were transplanted with FMT from long-term survival patients showed augmentation in tumor infiltration and activation of CD8+ T cells, upregulations of serum IFN-γ and IL-2 and decreased tumor burden, while mice that were transplanted with FMT from short-term survival patients showed enhancement in regulatory T cells and myeloid-derived suppressor cells in tumor infiltration and promoted tumor growth [236]. This indicates that gut microbiota from PDAC patients contributed to the modulation of tumor microbiome composition, anti-tumor immunity and tumor growth progression in treated mice.
The gut microbiota is able to have a distant effect as well on liver tumorigenesis via the transportation of pathogen metabolites, the associated molecular patterns and antigens from intestinal bacteria through the hepatic portal vein to the liver. It has been shown that secondary bile acids (BAs), such as lithocholic acid or murine-specific ω-muricholic acid, produced by Clostridium species presented pro-tumor effects in sorts of preclinical models of primary or metastatic liver cancer [237]. The mechanism was inferred to be related to the secondary BA-induced decrease in CXCL16 expression in liver sinusoidal endothelial cells, which attenuated recruitment of NK T cells to the liver. In fact, overproduction of the secondary BA deoxycholic acid could be due to the enrichment of Clostridium species in the gut caused by a high-fat diet, and deoxycholic acid accumulation resulted in the stimulation of the TLR2 pathway, leading to COX2 overexpression and, thereby, prostaglandin E2 (PGE2) overproduction in consequence [238]. PGE2 is shown to play a role in accelerating tumor progression through the suppression of tumor immunity [238]. Therefore, accumulation of the secondary BA deoxycholic acid is involved in pro-tumor effects on carcinogen-induced liver cancer.
It has been hypothesized that digestive tract microbiota may influence the biological functions and processes of all systems in the host through different signaling mechanisms. For example, the digestive tract microbiome may be involved in the modulation of sex hormone level through metabolizing and synthesizing and may signal to the central nervous system and potentially facilitate the progression of hormone-related cancers, including endometrial, ovarian, prostate and breast cancer [239,240,241]. It has been also shown that bacterial metabolites such as short-chain fatty acids, secondary Bas and enterolignans could influence tumor development and progression through the regulation of immune-mediated signals via host circulation access [242,243]. The gut microbiota is therefore regarded to constitute a reciprocal circuitry called the gut–brain axis [244,245]. Neuroinflammation induced by the gut microbiota could be contributing to the pathogenesis of diverse central nervous system diseases. An in vivo study showed that inhibition of the immunosuppressive effect of myeloid-derived suppressor cells caused the triggering of activation of the reward system of the brain in mice, which resulted in the alleviation of lung tumor growth [246]. This may indicate that the gut–brain axis could be a pathway via which the gut microbiota could influence the reward system through arousing immunosuppressive effects and contribute to cancer progression systemically [247]. Tumor development is intricately relevant to the host immune system, which is intimately influenced by the gut microbiota, leading to the potential involvement of the gut microbiota in tumor progression and therapeutic efficacy through direct intervening by bacterial metabolites or indirect alteration of immune homeostasis (Figure 6). This is evidently supported by preclinical and clinical studies in various cancers [234,235,248,249,250,251,252,253].

8. Conclusions

A good understanding of the mechanisms of cancer immunity could provide therapeutic opportunities. In general, the strategy includes shutting immune suppression and restoring antitumor immune responses through modulating the activities of the components involved in the immune landscape of cancer.
The synergistic action of both arms, innate and adaptive immunity, forms the basis of the carefully arranged anti-tumor response of the host immune system. The CTLs select for target killing by recognizing the tumor antigen peptides presented by MHC-I, subsequently killing tumor cells and providing support for anti-tumor immunity. MHC-I deficiency in cancers is commonly caused mostly by NLRC5 loss, which has been regarded as poor prognosis for many cancers. Restoring NLRC5 expression can be helpful for tumor immunogenicity and the recognition of cancer antigenic peptides. Although MHC-II, canonically expressed by APCs, has been shown to play multiple key roles in antitumor immunity, tsMHC-II, expressed on various tumor cells, exhibits a better prognostic correlation with cancer patients, both with patient survival and with response to immunotherapy with anti-PD-1/anti-PD-L1 drugs. TsMHC-II upregulation can augment cancer immunity and, thus, could be a novel strategy to ameliorate anti-tumor immune responses. TsMHC-II holds promise as a predictive biomarker of cancer immunotherapy compared with other parameters. NK cells are considered as crucial members of the complex immune response: in the primary immune response, via directly making lysis of cancer cells through the ADCC pathway and removing MHC-damaged cells; for the secondary adaptive immune response, by priming the tumor microenvironment via inducing PD-L1 expression on tumors and the recruitment of DCs and T cells. Therefore, without an NK cell compartment, it could be able to neither mount killing of malignant cells nor harness the full efficacy of immunotherapies. It is thus necessary to reconstitute the NK cell compartment via precursory adoptive NK cell transfer, which triggers the priming of the immune system for optimal effector populations’ activity and therapeutic efficacy. The differentiated γδ T cell population initially produces antitumor effects, which, however, can be restricted by their low proliferation and self-renewal capability, rendering their effect less persistent. In contrast, γδ T cells in naive or central memory status have long persistence and are amenable to continuous tumor surveillance. After antigen stimulation, this cell population can differentiate into effector cells that recognize tumors and then retain a subpopulation of self-renewing memory cells for subsequent tumor encounters and corresponding responses. Therapeutically, naïve γδ T cells or those in central memory status are applicable for clinical use, but generating a large enough population in vitro for therapeutic desire could still be challenging. It is suggested by the available data that eosinophils are involved in the facilitation of tumor immunity via cross-talk with various lymphocyte subsets directly and indirectly. But, the transcriptomic profile and individual heterogeneity of eosinophils are far from being elucidated, for eosinophils are often undetectable by most single-cell RNA-sequencing analyses [254], rendering us limited to fully elucidate unique eosinophil subsets, eosinophils interacting with specific subsets in the tumor microenvironment or their distinct functions. New experimental approaches are required to overcome these challenges. Commensal microbiota, via their pronounced impact on host immunity, have been shown to influence anti-tumor immunity and therapeutic efficacy. Clinically, they are mainly based on the application of microbial agents with immunostimulatory effects to enhance the efficacy of cancer immunotherapy. Further understanding microbe-mediated immunoregulatory pathways and components, and discovering strains with precise immunostimulatory and immunosuppressive effects, may contribute to facilitating precise and personal therapeutic approaches that may render fewer undesirable outcomes. In addition, it is plausible to inhibit the proliferation and decrease the abundance of immune-suppressing or tumor-promoting bacteria, which may enhance immunotherapeutic efficacy.

Author Contributions

Conceptualization, Q.W.; writing—original draft preparation, Y.L., X.Q. and Q.W.; writing—review and editing, M.W., B.Q. and Q.W.; visualization, supervision, Q.W.; project administration, funding acquisition, M.W. and Q.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Intramural scientific research project of Guangzhou University with grant number [RP2021032], Guangzhou Municipal-school (college) Jointly Funded the Basic and Applied Basic Research Projects with grant number [202201020224], and Innovation and Entrepreneurship Training Programs for College Students in Guangzhou University [202111078036].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zou, W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat. Rev. Cancer 2005, 5, 263–274. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Joyce, J.A.; Fearon, D.T. T cell exclusion, immune privilege, and the tumor microenvironment. Science 2015, 348, 74–80. [Google Scholar] [CrossRef] [Green Version]
  4. Parham, P.; Lomen, C.E.; Lawlor, D.A.; Ways, J.P.; Holmes, N.; Coppin, H.L.; Salter, R.D.; Wan, A.M.; Ennis, P.D. Nature of polymorphism in HLA-A, -B, and -C molecules. Proc. Natl. Acad. Sci. USA 1988, 85, 4005–4009. [Google Scholar] [CrossRef] [PubMed]
  5. Bjorkman, P.J.; Parham, P. Structure, function, and diversity of class I major histocompatibility complex molecules. Annu. Rev. Biochem. 1990, 59, 253–288. [Google Scholar] [CrossRef] [PubMed]
  6. Shawar, S.M.; Vyas, J.M.; Rodgers, J.R.; Rich, R.R. Antigen presentation by major histocompatibility complex class I-B molecules. Annu. Rev. Immunol. 1994, 12, 839–880. [Google Scholar] [CrossRef]
  7. D’Souza, M.P.; Adams, E.; Altman, J.D.; Birnbaum, M.E.; Boggiano, C.; Casorati, G.; Chien, Y.H.; Conley, A.; Eckle, S.B.G.; Fruh, K.; et al. Casting a wider net: Immunosurveillance by nonclassical MHC molecules. PLoS Pathog. 2019, 15, e1007567. [Google Scholar] [CrossRef] [Green Version]
  8. Hicklin, D.J.; Marincola, F.M.; Ferrone, S. HLA class I antigen downregulation in human cancers: T-cell immunotherapy revives an old story. Mol. Med. Today 1999, 5, 178–186. [Google Scholar] [CrossRef]
  9. Koopman, L.A.; Corver, W.E.; van der Slik, A.R.; Giphart, M.J.; Fleuren, G.J. Multiple genetic alterations cause frequent and heterogeneous human histocompatibility leukocyte antigen class I loss in cervical cancer. J. Exp. Med. 2000, 191, 961–976. [Google Scholar] [CrossRef]
  10. Paschen, A.; Mendez, R.M.; Jimenez, P.; Sucker, A.; Ruiz-Cabello, F.; Song, M.; Garrido, F.; Schadendorf, D. Complete loss of HLA class I antigen expression on melanoma cells: A result of successive mutational events. Int. J. Cancer 2003, 103, 759–767. [Google Scholar] [CrossRef]
  11. Bukur, J.; Jasinski, S.; Seliger, B. The role of classical and non-classical HLA class I antigens in human tumors. Semin. Cancer Biol. 2012, 22, 350–358. [Google Scholar] [CrossRef] [PubMed]
  12. Sokol, L.; Koelzer, V.H.; Rau, T.T.; Karamitopoulou, E.; Zlobec, I.; Lugli, A. Loss of tapasin correlates with diminished CD8(+) T-cell immunity and prognosis in colorectal cancer. J. Transl. Med. 2015, 13, 279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Campoli, M.; Chang, C.C.; Ferrone, S. HLA class I antigen loss, tumor immune escape and immune selection. Vaccine 2002, 20 (Suppl. S4), A40–A45. [Google Scholar] [CrossRef] [PubMed]
  14. Cabrera, T.; Lara, E.; Romero, J.M.; Maleno, I.; Real, L.M.; Ruiz-Cabello, F.; Valero, P.; Camacho, F.M.; Garrido, F. HLA class I expression in metastatic melanoma correlates with tumor development during autologous vaccination. Cancer Immunol. Immunother. CII 2007, 56, 709–717. [Google Scholar] [CrossRef] [PubMed]
  15. Seliger, B.; Ruiz-Cabello, F.; Garrido, F. IFN inducibility of major histocompatibility antigens in tumors. Adv. Cancer Res. 2008, 101, 249–276. [Google Scholar]
  16. Campoli, M.; Ferrone, S. HLA antigen changes in malignant cells: Epigenetic mechanisms and biologic significance. Oncogene 2008, 27, 5869–5885. [Google Scholar] [CrossRef] [Green Version]
  17. 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]
  18. 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]
  19. Cui, J.; Zhu, L.; Xia, X.; Wang, H.Y.; Legras, X.; Hong, J.; Ji, J.; Shen, P.; Zheng, S.; Chen, Z.J.; et al. NLRC5 negatively regulates the NF-kappaB and type I interferon signaling pathways. Cell 2010, 141, 483–496. [Google Scholar] [CrossRef] [Green Version]
  20. Benko, S.; Magalhaes, J.G.; Philpott, D.J.; Girardin, S.E. NLRC5 limits the activation of inflammatory pathways. J. Immunol. 2010, 185, 1681–1691. [Google Scholar] [CrossRef] [Green Version]
  21. Staehli, F.; Ludigs, K.; Heinz, L.X.; Seguin-Estevez, 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] [Green Version]
  22. Kuenzel, S.; Till, A.; Winkler, M.; Hasler, R.; Lipinski, S.; Jung, S.; Grotzinger, 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] [Green Version]
  23. Neerincx, A.; Rodriguez, G.M.; Steimle, V.; Kufer, T.A. NLRC5 controls basal MHC class I gene expression in an MHC enhanceosome-dependent manner. J. Immunol. 2012, 188, 4940–4950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Robbins, G.R.; Truax, A.D.; Davis, B.K.; Zhang, L.; Brickey, W.J.; Ting, J.P. Regulation of class I major histocompatibility complex (MHC) by nucleotide-binding domain, leucine-rich repeat-containing (NLR) proteins. J. Biol. Chem. 2012, 287, 24294–24303. [Google Scholar] [CrossRef] [Green Version]
  25. Lupfer, C.R.; Stokes, K.L.; Kuriakose, T.; Kanneganti, T.D. Deficiency of the NOD-Like Receptor NLRC5 Results in Decreased CD8(+) T Cell Function and Impaired Viral Clearance. J. Virol. 2017, 91, e00377-17. [Google Scholar] [CrossRef] [Green Version]
  26. Sun, T.; Ferrero, R.L.; Girardin, S.E.; Gommerman, J.L.; Philpott, D.J. NLRC5 deficiency has a moderate impact on immunodominant CD8(+) T cell responses during rotavirus infection of adult mice. Immunol. Cell Biol. 2019, 97, 552–562. [Google Scholar] [CrossRef]
  27. Kobayashi, K.S. NLRC5/CITA: A novel regulator of class I major histocompatibility complex genes. J. Immunodefic. Disord. 2012, 1, 1000e102. [Google Scholar] [CrossRef] [PubMed]
  28. Biswas, A.; Meissner, T.B.; Kawai, T.; Kobayashi, K.S. Cutting edge: Impaired MHC class I expression in mice deficient for Nlrc5/class I transactivator. J. Immunol. 2012, 189, 516–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Yao, Y.; Wang, Y.; Chen, F.; Huang, Y.; Zhu, S.; Leng, Q.; Wang, H.; Shi, Y.; Qian, Y. NLRC5 regulates MHC class I antigen presentation in host defense against intracellular pathogens. Cell Res. 2012, 22, 836–847. [Google Scholar] [CrossRef] [Green Version]
  30. Dang, A.T.; Strietz, J.; Zenobi, A.; Khameneh, H.J.; Brandl, S.M.; Lozza, L.; Conradt, G.; Kaufmann, S.H.E.; Reith, W.; Kwee, I.; et al. NLRC5 promotes transcription of BTN3A1-3 genes and Vgamma9Vdelta2 T cell-mediated killing. iScience 2021, 24, 101900. [Google Scholar] [CrossRef]
  31. Zhang, P.; Yu, C.; Yu, J.; Li, Z.; Lan, H.Y.; Zhou, Q. Arid2-IR promotes NF-kappaB-mediated renal inflammation by targeting NLRC5 transcription. Cell Mol. Life Sci. 2020, 78, 2387–2404. [Google Scholar] [CrossRef]
  32. Zhou, Q.; Huang, X.R.; Yu, J.; Yu, X.; Lan, H.Y. Long Noncoding RNA Arid2-IR Is a Novel Therapeutic Target for Renal Inflammation. Mol. Ther. J. Am. Soc. Gene Ther. 2015, 23, 1034–1043. [Google Scholar] [CrossRef] [Green Version]
  33. Zong, Z.; Song, Y.; Xue, Y.; Ruan, X.; Liu, X.; Yang, C.; Zheng, J.; Cao, S.; Li, Z.; Liu, Y. Knockdown of LncRNA SCAMP1 suppressed malignant biological behaviours of glioma cells via modulating miR-499a-5p/LMX1A/NLRC5 pathway. J. Cell Mol. Med. 2019, 23, 5048–5062. [Google Scholar] [CrossRef] [Green Version]
  34. Periyasamy, P.; Thangaraj, A.; Bendi, V.S.; Buch, S. HIV-1 Tat-mediated microglial inflammation involves a novel miRNA-34a-NLRC5-NFkappaB signaling axis. Brain Behav. Immun. 2019, 80, 227–237. [Google Scholar] [CrossRef]
  35. Li, J.; Yu, L.; Shen, Z.; Li, Y.; Chen, B.; Wei, W.; Chen, X.; Wang, Q.; Tong, F.; Lou, H.; et al. miR-34a and its novel target, NLRC5, are associated with HPV16 persistence. Infect. Genet. Evol. 2016, 44, 293–299. [Google Scholar] [CrossRef]
  36. Zong, Y.; Zhang, Y.; Hou, D.; Xu, J.; Cui, F.; Qin, Y.; Sun, X. The lncRNA XIST promotes the progression of breast cancer by sponging miR-125b-5p to modulate NLRC5. Am. J. Transl. Res. 2020, 12, 3501–3511. [Google Scholar]
  37. Agudo, J.; Park, E.S.; Rose, S.A.; Alibo, E.; Sweeney, R.; Dhainaut, M.; Kobayashi, K.S.; Sachidanandam, R.; Baccarini, A.; Merad, M.; et al. Quiescent Tissue Stem Cells Evade Immune Surveillance. Immunity 2018, 48, 271–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Schatton, T.; Schutte, U.; Frank, N.Y.; Zhan, Q.; Hoerning, A.; Robles, S.C.; Zhou, J.; Hodi, F.S.; Spagnoli, G.C.; Murphy, G.F.; et al. Modulation of T-cell activation by malignant melanoma initiating cells. Cancer Res. 2010, 70, 697–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Di Tomaso, T.; Mazzoleni, S.; Wang, E.; Sovena, G.; Clavenna, D.; Franzin, A.; Mortini, P.; Ferrone, S.; Doglioni, C.; Marincola, F.M.; et al. Immunobiological characterization of cancer stem cells isolated from glioblastoma patients. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010, 16, 800–813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Maccalli, C.; Volonte, A.; Cimminiello, C.; Parmiani, G. Immunology of cancer stem cells in solid tumours. A review. Eur. J. Cancer 2014, 50, 649–655. [Google Scholar] [CrossRef] [PubMed]
  41. Bruttel, V.S.; Wischhusen, J. Cancer stem cell immunology: Key to understanding tumorigenesis and tumor immune escape? Front. Immunol. 2014, 5, 360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Sultan, M.; Coyle, K.M.; Vidovic, D.; Thomas, M.L.; Gujar, S.; Marcato, P. Hide-and-seek: The interplay between cancer stem cells and the immune system. Carcinogenesis 2017, 38, 107–118. [Google Scholar] [CrossRef] [PubMed]
  43. Morrison, B.J.; Steel, J.C.; Morris, J.C. Reduction of MHC-I expression limits T-lymphocyte-mediated killing of Cancer-initiating cells. BMC Cancer 2018, 18, 469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. 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, e1151593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Seliger, B.; Wollscheid, U.; Momburg, F.; Blankenstein, T.; Huber, C. Characterization of the major histocompatibility complex class I deficiencies in B16 melanoma cells. Cancer Res. 2001, 61, 1095–1099. [Google Scholar]
  46. Kim, H.; Kim, H.; Feng, Y.; Li, Y.; Tamiya, H.; Tocci, S.; Ronai, Z.A. PRMT5 control of cGAS/STING and NLRC5 pathways defines melanoma response to antitumor immunity. Sci. Transl. Med. 2020, 12, eaaz5683. [Google Scholar] [CrossRef]
  47. Zebertavage, L.K.; Alice, A.; Crittenden, M.R.; Gough, M.J. Transcriptional Upregulation of NLRC5 by Radiation Drives STINGand Interferon-Independent MHC-I Expression on Cancer Cells and T Cell Cytotoxicity. Sci. Rep. 2020, 10, 7376. [Google Scholar] [CrossRef]
  48. Kalbasi, A.; Tariveranmoshabad, M.; Hakimi, K.; Kremer, S.; Campbell, K.M.; Funes, J.M.; Vega-Crespo, A.; Parisi, G.; Champekar, A.; Nguyen, C.; et al. Uncoupling interferon signaling and antigen presentation to overcome immunotherapy resistance due to JAK1 loss in melanoma. Sci. Transl. Med. 2020, 12, eabb0152. [Google Scholar] [CrossRef]
  49. Peng, Y.Y.; He, Y.H.; Chen, C.; Xu, T.; Li, L.; Ni, M.M.; Meng, X.M.; Huang, C.; Li, J. NLRC5 regulates cell proliferation, migration and invasion in hepatocellular carcinoma by targeting the Wnt/beta-catenin signaling pathway. Cancer Lett. 2016, 376, 10–21. [Google Scholar] [CrossRef] [Green Version]
  50. Wang, Q.; Ding, H.; He, Y.; Li, X.; Cheng, Y.; Xu, Q.; Yang, Y.; Liao, G.; Meng, X.; Huang, C.; et al. NLRC5 mediates cell proliferation, migration, and invasion by regulating the Wnt/beta-catenin signalling pathway in clear cell renal cell carcinoma. Cancer Lett. 2019, 444, 9–19. [Google Scholar] [CrossRef]
  51. He, Y.H.; Li, M.F.; Zhang, X.Y.; Meng, X.M.; Huang, C.; Li, J. NLRC5 promotes cell proliferation via regulating the AKT/VEGF-A signaling pathway in hepatocellular carcinoma. Toxicology 2016, 359–360, 47–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Fan, Y.; Dong, Z.; Shi, Y.; Sun, S.; Wei, B.; Zhan, L. NLRC5 promotes cell migration and invasion by activating the PI3K/AKT signaling pathway in endometrial cancer. J. Int. Med. Res. 2020, 48, 300060520925352. [Google Scholar] [CrossRef] [PubMed]
  53. Hu, X.; Wang, M.; Cao, L.; Cong, L.; Gao, Y.; Lu, J.; Feng, J.; Shen, B.; Liu, D. miR-4319 Suppresses the Growth of Esophageal Squamous Cell Carcinoma Via Targeting NLRC5. Curr. Mol. Pharm. 2020, 13, 144–149. [Google Scholar] [CrossRef] [PubMed]
  54. Arnett, H.A.; Viney, J.L. Immune modulation by butyrophilins. Nat. Rev. Immunol. 2014, 14, 559–569. [Google Scholar] [CrossRef]
  55. Mayor, A.; Martinon, F.; De Smedt, T.; Petrilli, V.; Tschopp, J. A crucial function of SGT1 and HSP90 in inflammasome activity links mammalian and plant innate immune responses. Nat. Immunol. 2007, 8, 497–503. [Google Scholar] [CrossRef]
  56. Budhwani, M.; Mazzieri, R.; Dolcetti, R. Plasticity of Type I Interferon-Mediated Responses in Cancer Therapy: From Anti-tumor Immunity to Resistance. Front. Oncol. 2018, 8, 322. [Google Scholar] [CrossRef] [Green Version]
  57. Xi, S.; Dyer, K.F.; Kimak, M.; Zhang, Q.; Gooding, W.E.; Chaillet, J.R.; Chai, R.L.; Ferrell, R.E.; Zamboni, B.; Hunt, J.; et al. Decreased STAT1 expression by promoter methylation in squamous cell carcinogenesis. J. Natl. Cancer Inst. 2006, 98, 181–189. [Google Scholar] [CrossRef] [Green Version]
  58. Komyod, W.; Bohm, M.; Metze, D.; Heinrich, P.C.; Behrmann, I. Constitutive suppressor of cytokine signaling 3 expression confers a growth advantage to a human melanoma cell line. Mol. Cancer Res. 2007, 5, 271–281. [Google Scholar] [CrossRef] [Green Version]
  59. Cheng, Y.; He, C.; Wang, M.; Ma, X.; Mo, F.; Yang, S.; Han, J.; Wei, X. Targeting epigenetic regulators for cancer therapy: Mechanisms and advances in clinical trials. Signal. Transduct Target. Ther. 2019, 4, 62. [Google Scholar] [CrossRef] [Green Version]
  60. Kungulovski, G.; Jeltsch, A. Epigenome Editing: State of the Art, Concepts, and Perspectives. Trends Genet. 2016, 32, 101–113. [Google Scholar] [CrossRef]
  61. Nebbioso, A.; Tambaro, F.P.; Dell’Aversana, C.; Altucci, L. Cancer epigenetics: Moving forward. PLoS Genet. 2018, 14, e1007362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Janssen, E.M.; Lemmens, E.E.; Wolfe, T.; Christen, U.; von Herrath, M.G.; Schoenberger, S.P. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 2003, 421, 852–856. [Google Scholar] [CrossRef] [PubMed]
  63. Sun, J.C.; Bevan, M.J. Defective CD8 T Cell Memory Following Acute Infection Without CD4 T Cell Help. Science 2003, 300, 339–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Laidlaw, B.J.; Craft, J.E.; Kaech, S.M. The multifaceted role of CD4+ T cells in CD8+ T cell memory. Nat. Rev. Immunol. 2016, 16, 102–111. [Google Scholar] [CrossRef] [Green Version]
  65. Ossendorp, F.; Mengedé, E.; Camps, M.; Filius, R.; Melief, C.J. Specific T helper cell requirement for optimal induction of cytotoxic T lymphocytes against major histocompatibility complex class II negative tumors. J. Exp. Med. 1998, 187, 693–702. [Google Scholar] [CrossRef]
  66. Spitzer, M.H.; Carmi, Y.; Reticker-Flynn, N.E.; Kwek, S.S.; Madhireddy, D.; Martins, M.M.; Gherardini, P.F.; Prestwood, T.R.; Chabon, J.; Bendall, S.C.; et al. Systemic Immunity Is Required for Effective Cancer Immunotherapy. Cell 2017, 168, 487–502. [Google Scholar] [CrossRef] [Green Version]
  67. Ahrends, T.; Babala, N.; Xiao, Y.; Yagita, H.; van Eenennaam, H.; Borst, J. CD27 Agonism Plus PD-1 Blockade Recapitulates CD4+ T-cell Help in Therapeutic Anticancer Vaccination. Cancer Res. 2016, 76, 2921–2931. [Google Scholar] [CrossRef] [Green Version]
  68. Quezada, S.A.; Simpson, T.R.; Peggs, K.S.; Merghoub, T.; Vider, J.; Fan, X.; Blasberg, R.; Yagita, H.; Muranski, P.; Antony, P.A.; et al. Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J. Exp. Med. 2010, 207, 637–650. [Google Scholar] [CrossRef]
  69. Bos, R.; Sherman, L.A. CD4+ T-Cell Help in the Tumor Milieu Is Required for Recruitment and Cytolytic Function of CD8+ T Lymphocytes. Cancer Res. 2010, 70, 8368–8377. [Google Scholar] [CrossRef] [Green Version]
  70. Homet Moreno, B.; Zaretsky, J.M.; Garcia-Diaz, A.; Tsoi, J.; Parisi, G.; Robert, L.; Meeth, K.; Ndoye, A.; Bosenberg, M.; Weeraratna, A.T.; et al. Response to Programmed Cell Death-1 Blockade in a Murine Melanoma Syngeneic Model Requires Costimulation, CD4, and CD8 T Cells. Cancer Immunol. Res. 2016, 4, 845–857. [Google Scholar] [CrossRef] [Green Version]
  71. Kambayashi, T.; Laufer, T.M. Atypical MHC class II-expressing antigen-presenting cells: Can anything replace a dendritic cell? Nat. Rev. Immunol. 2014, 14, 719–730. [Google Scholar] [CrossRef] [PubMed]
  72. Johnson, D.B.; Estrada, M.V.; Salgado, R.; Sanchez, V.; Doxie, D.B.; Opalenik, S.R.; Vilgelm, A.E.; Feld, E.; Johnson, A.S.; Greenplate, A.R.; et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat. Commun. 2016, 7, 10582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Forero, A.; Li, Y.; Chen, D.; Grizzle, W.E.; Updike, K.L.; Merz, N.D.; Downs-Kelly, E.; Burwell, T.C.; Vaklavas, C.; Buchsbaum, D.J.; et al. Expression of the MHC Class II Pathway in Triple-Negative Breast Cancer Tumor Cells Is Associated with a Good Prognosis and Infiltrating Lymphocytes. Cancer Immunol. Res. 2016, 4, 390–399. [Google Scholar] [CrossRef] [Green Version]
  74. Park, I.A.; Hwang, S.H.; Song, I.H.; Heo, S.H.; Kim, Y.A.; Bang, W.S.; Park, H.S.; Lee, M.; Gong, G.; Lee, H.J. Expression of the MHC class II in triple-negative breast cancer is associated with tumor-infiltrating lymphocytes and interferon signaling. PLoS ONE 2017, 12, e0182786. [Google Scholar] [CrossRef] [Green Version]
  75. Armstrong, T.D.; Clements, V.K.; Martin, B.K.; Ting, J.P.; Ostrand-Rosenberg, S. Major histocompatibility complex class II-transfected tumor cells present endogenous antigen and are potent inducers of tumor-specific immunity. Proc. Natl. Acad. Sci. USA 1997, 94, 6886–6891. [Google Scholar] [CrossRef] [PubMed]
  76. Roemer, M.G.M.; Redd, R.A.; Cader, F.Z.; Pak, C.J.; Abdelrahman, S.; Ouyang, J.; Sasse, S.; Younes, A.; Fanale, M.; Santoro, A.; et al. Major Histocompatibility Complex Class II and Programmed Death Ligand 1 Expression Predict Outcome After Programmed Death 1 Blockade in Classic Hodgkin Lymphoma. J. Clin. Oncol. 2018, 36, 942–950. [Google Scholar] [CrossRef]
  77. Rodig, S.J.; Gusenleitner, D.; Jackson, D.G.; Gjini, E.; Giobbie-Hurder, A.; Jin, C.; Chang, H.; Lovitch, S.B.; Horak, C.; Weber, J.S.; et al. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci. Transl. Med. 2018, 10, eaar3342. [Google Scholar] [CrossRef] [Green Version]
  78. Trowsdale, J. Genomic structure and function in the MHC. Trends Genet. 1993, 9, 117–122. [Google Scholar] [CrossRef]
  79. Unanue, E.R.; Turk, V.; Neefjes, J. Variations in MHC Class II Antigen Processing and Presentation in Health and Disease. Annu. Rev. Immunol. 2016, 34, 265–297. [Google Scholar] [CrossRef]
  80. Arnold, P.Y.; La Gruta, N.L.; Miller, T.; Vignali, K.M.; Adams, P.S.; Woodland, D.L.; Vignali, D.A. The majority of immunogenic epitopes generate CD4+ T cells that are dependent on MHC class II-bound peptide-flanking residues. J. Immunol. 2002, 169, 739–749. [Google Scholar] [CrossRef]
  81. Mortara, L.; Castellani, P.; Meazza, R.; Tosi, G.; De Lerma Barbaro, A.; Procopio, F.A.; Comes, A.; Zardi, L.; Ferrini, S.; Accolla, R.S. CIITA-induced MHC class II expression in mammary adenocarcinoma leads to a Th1 polarization of the tumor microenvironment, tumor rejection, and specific antitumor memory. Clin. Cancer. Res. 2006, 12, 3435–3443. [Google Scholar] [CrossRef] [Green Version]
  82. Baskar, S.; Clements, V.K.; Glimcher, L.H.; Nabavi, N.; Ostrand-Rosenberg, S. Rejection of MHC class II-transfected tumor cells requires induction of tumor-encoded B7-1 and/or B7-2 costimulatory molecules. J. Immunol. 1996, 156, 3821–3827. [Google Scholar] [CrossRef]
  83. Loi, S.; Dushyanthen, S.; Beavis, P.A.; Salgado, R.; Denkert, C.; Savas, P.; Combs, S.; Rimm, D.L.; Giltnane, J.M.; Estrada, M.V.; et al. RAS/MAPK Activation Is Associated with Reduced Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Therapeutic Cooperation Between MEK and PD-1/PD-L1 Immune Checkpoint Inhibitors. Clin. Cancer. Res. 2016, 22, 1499–1509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Concha, A.; Ruiz-Cabello, F.; Cabrera, T.; Nogales, F.; Collado, A.; Garrido, F. Different patterns of HLA-DR antigen expression in normal epithelium, hyperplastic and neoplastic malignant lesions of the breast. Eur. J. Immunogenet. 1995, 22, 299–310. [Google Scholar] [CrossRef] [PubMed]
  85. Oldford, S.A.; Robb, J.D.; Codner, D.; Gadag, V.; Watson, P.H.; Drover, S. Tumor cell expression of HLA-DM associates with a Th1 profile and predicts improved survival in breast carcinoma patients. Int. Immunol. 2006, 18, 1591–1602. [Google Scholar] [CrossRef] [Green Version]
  86. Oldford, S.A.; Robb, J.D.; Watson, P.H.; Drover, S. HLA-DRB alleles are differentially expressed by tumor cells in breast carcinoma. Int. J. Cancer 2004, 112, 399–406. [Google Scholar] [CrossRef] [PubMed]
  87. Jabrane-Ferrat, N.; Faille, A.; Loiseau, P.; Poirier, O.; Charron, D.; Calvo, F. Effect of gamma interferon on HLA class-I and -II transcription and protein expression in human breast adenocarcinoma cell lines. Int. J. Cancer 1990, 45, 1169–1176. [Google Scholar] [CrossRef] [PubMed]
  88. da Silva, G.B.; Silva, T.G.; Duarte, R.A.; Neto, N.L.; Carrara, H.H.; Donadi, E.A.; Gonçalves, M.A.; Soares, E.G.; Soares, C.P. Expression of the Classical and Nonclassical HLA Molecules in Breast Cancer. Int. J. Breast Cancer 2013, 2013, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Feinmesser, M.; Sulkes, A.; Morgenstern, S.; Sulkes, J.; Stern, S.; Okon, E. HLA-DR and beta 2 microglobulin expression in medullary and atypical medullary carcinoma of the breast: Histopathologically similar but biologically distinct entities. J. Clin. Pathol. 2000, 53, 286–291. [Google Scholar] [CrossRef] [Green Version]
  90. Bártek, J.; Petrek, M.; Vojtĕsek, B.; Bártková, J.; Kovarík, J.; Rejthar, A. HLA-DR antigens on differentiating human mammary gland epithelium and breast tumours. Br. J. Cancer 1987, 56, 727–733. [Google Scholar] [CrossRef] [Green Version]
  91. Michel, S.; Linnebacher, M.; Alcaniz, J.; Voss, M.; Wagner, R.; Dippold, W.; Becker, C.; von Knebel Doeberitz, M.; Ferrone, S.; Kloor, M. Lack of HLA class II antigen expression in microsatellite unstable colorectal carcinomas is caused by mutations in HLA class II regulatory genes. Int. J. Cancer 2010, 127, 889–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Bustin, S.A.; Li, S.-R.; Phillips, S.; Dorudi, S. Expression of HLA Class II in Colorectal Cancer: Evidence for Enhanced Immunogenicity of Microsatellite-Instability-Positive Tumours. Tumor Biol. 2001, 22, 294–298. [Google Scholar] [CrossRef] [PubMed]
  93. Soos, J.M.; Krieger, J.I.; Stüve, O.; King, C.L.; Patarroyo, J.C.; Aldape, K.; Wosik, K.; Slavin, A.J.; Nelson, P.A.; Antel, J.P.; et al. Malignant glioma cells use MHC class II transactivator (CIITA) promoters III and IV to direct IFN-gamma-inducible CIITA expression and can function as nonprofessional antigen presenting cells in endocytic processing and CD4(+) T-cell activation. Glia 2001, 36, 391–405. [Google Scholar] [CrossRef] [PubMed]
  94. Johnson, D.B.; Bordeaux, J.; Kim, J.Y.; Vaupel, C.; Rimm, D.L.; Ho, T.H.; Joseph, R.W.; Daud, A.I.; Conry, R.M.; Gaughan, E.M.; et al. Quantitative Spatial Profiling of PD-1/PD-L1 Interaction and HLA-DR/IDO-1 Predicts Improved Outcomes of anti-PD-1 Therapies in Metastatic Melanoma. Clin. Cancer Res. 2018, 24, 5250–5260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Yazawa, T.; Kamma, H.; Fujiwara, M.; Matsui, M.; Horiguchi, H.; Satoh, H.; Fujimoto, M.; Yokoyama, K.; Ogata, T. Lack of class II transactivator causes severe deficiency of HLA-DR expression in small cell lung cancer. J. Pathol. 1999, 187, 191–199. [Google Scholar] [CrossRef]
  96. Callahan, M.J.; Nagymanyoki, Z.; Bonome, T.; Johnson, M.E.; Litkouhi, B.; Sullivan, E.H.; Hirsch, M.S.; Matulonis, U.A.; Liu, J.; Birrer, M.J.; et al. Increased HLA-DMB Expression in the Tumor Epithelium Is Associated with Increased CTL Infiltration and Improved Prognosis in Advanced-Stage Serous Ovarian Cancer. Clin. Cancer Res. 2008, 14, 7667–7673. [Google Scholar] [CrossRef] [Green Version]
  97. Turner, T.B.; Meza-Perez, S.; Londoño, A.; Katre, A.; Peabody, J.E.; Smith, H.J.; Forero, A.; Norian, L.A.; Straughn, J.M., Jr.; Buchsbaum, D.J.; et al. Epigenetic modifiers upregulate MHC II and impede ovarian cancer tumor growth. Oncotarget 2017, 8, 44159–44170. [Google Scholar] [CrossRef] [Green Version]
  98. Younger, A.R.; Amria, S.; Jeffrey, W.A.; Mahdy, A.E.; Goldstein, O.G.; Norris, J.S.; Haque, A. HLA class II antigen presentation by prostate cancer cells. Prostate Cancer Prostatic Dis. 2008, 11, 334–341. [Google Scholar] [CrossRef] [Green Version]
  99. Kim, H.-J.; Cantor, H. CD4 T-cell Subsets and Tumor Immunity: The Helpful and the Not-so-Helpful. Cancer Immunol. Res. 2014, 2, 91–98. [Google Scholar] [CrossRef] [Green Version]
  100. Linnemann, C.; van Buuren, M.M.; Bies, L.; Verdegaal, E.M.; Schotte, R.; Calis, J.J.; Behjati, S.; Velds, A.; Hilkmann, H.; Atmioui, D.E.; et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat. Med. 2015, 21, 81–85. [Google Scholar] [CrossRef]
  101. Kreiter, S.; Vormehr, M.; van de Roemer, N.; Diken, M.; Löwer, M.; Diekmann, J.; Boegel, S.; Schrörs, B.; Vascotto, F.; Castle, J.C.; et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015, 520, 692–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Dadmarz, R.; Sgagias, M.K.; Rosenberg, S.A.; Schwartzentruber, D.J. CD4+ T lymphocytes infiltrating human breast cancer recognise autologous tumor in an MHC-class-II restricted fashion. Cancer Immunol. Immunother. 1995, 40, 1–9. [Google Scholar] [PubMed]
  103. Levine, A.G.; Arvey, A.; Jin, W.; Rudensky, A.Y. Continuous requirement for the TCR in regulatory T cell function. Nat. Immunol. 2014, 15, 1070–1078. [Google Scholar] [CrossRef] [Green Version]
  104. Plitas, G.; Konopacki, C.; Wu, K.; Bos, P.D.; Morrow, M.; Putintseva, E.V.; Chudakov, D.M.; Rudensky, A.Y. Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity 2016, 45, 1122–1134. [Google Scholar] [CrossRef] [Green Version]
  105. Jang, J.E.; Hajdu, C.H.; Liot, C.; Miller, G.; Dustin, M.L.; Bar-Sagi, D. Crosstalk between Regulatory T Cells and Tumor-Associated Dendritic Cells Negates Anti-tumor Immunity in Pancreatic Cancer. Cell Rep. 2017, 20, 558–571. [Google Scholar] [CrossRef] [Green Version]
  106. Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242. [Google Scholar] [CrossRef]
  107. Driessens, G.; Kline, J.; Gajewski, T.F. Costimulatory and coinhibitory receptors in anti-tumor immunity. Immunol. Rev. 2009, 229, 126–144. [Google Scholar] [CrossRef]
  108. Haabeth, O.A.; Tveita, A.A.; Fauskanger, M.; Schjesvold, F.; Lorvik, K.B.; Hofgaard, P.O.; Omholt, H.; Munthe, L.A.; Dembic, Z.; Corthay, A.; et al. How Do CD4(+) T Cells Detect and Eliminate Tumor Cells That Either Lack or Express MHC Class II Molecules? Front. Immunol. 2014, 5, 174. [Google Scholar] [CrossRef] [PubMed]
  109. Jilaveanu, L.B.; Sznol, J.; Aziz, S.A.; Duchen, D.; Kluger, H.M.; Camp, R.L. CD70 expression patterns in renal cell carcinoma. Hum. Pathol. 2012, 43, 1394–1399. [Google Scholar] [CrossRef] [Green Version]
  110. Shibahara, I.; Saito, R.; Zhang, R.; Chonan, M.; Shoji, T.; Kanamori, M.; Sonoda, Y.; Kumabe, T.; Kanehira, M.; Kikuchi, T.; et al. OX40 ligand expressed in glioblastoma modulates adaptive immunity depending on the microenvironment: A clue for successful immunotherapy. Mol. Cancer 2015, 14, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Sartoris, S.; Valle, M.T.; Barbaro, A.L.; Tosi, G.; Cestari, T.; D’Agostino, A.; Megiovanni, A.M.; Manca, F.; Accolla, R.S. HLA class II expression in uninducible hepatocarcinoma cells after transfection of AIR-1 gene product CIITA: Acquisition of antigen processing and presentation capacity. J. Immunol. 1998, 161, 814–820. [Google Scholar] [CrossRef] [PubMed]
  112. Steimle, V.; Siegrist, C.A.; Mottet, A.; Lisowska-Grospierre, B.; Mach, B. Regulation of MHC class II expression by interferon-gamma mediated by the transactivator gene CIITA. Science 1994, 265, 106–109. [Google Scholar] [CrossRef] [PubMed]
  113. Chang, C.H.; Fontes, J.D.; Peterlin, M.; Flavell, R.A. Class II transactivator (CIITA) is sufficient for the inducible expression of major histocompatibility complex class II genes. J. Exp. Med. 1994, 180, 1367–1374. [Google Scholar] [CrossRef] [PubMed]
  114. Meraz, M.A.; White, J.M.; Sheehan, K.C.; Bach, E.A.; Rodig, S.J.; Dighe, A.S.; Kaplan, D.H.; Riley, J.K.; Greenlund, A.C.; Campbell, D.; et al. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK-STAT signaling pathway. Cell 1996, 84, 431–442. [Google Scholar] [CrossRef] [Green Version]
  115. Martins, I.; Deshayes, F.; Baton, F.; Forget, A.; Ciechomska, I.; Sylla, K.; Aoudjit, F.; Charron, D.; Al-Daccak, R.; Alcaide-Loridan, C. Pathologic expression of MHC class II is driven by mitogen-activated protein kinases. Eur. J. Immunol. 2007, 37, 788–797. [Google Scholar] [CrossRef]
  116. Mottok, A.; Woolcock, B.; Chan, F.C.; Tong, K.M.; Chong, L.; Farinha, P.; Telenius, A.; Chavez, E.; Ramchandani, S.; Drake, M.; et al. Genomic Alterations in CIITA Are Frequent in Primary Mediastinal Large B Cell Lymphoma and Are Associated with Diminished MHC Class II Expression. Cell Rep. 2015, 13, 1418–1431. [Google Scholar] [CrossRef] [Green Version]
  117. Steidl, C.; Shah, S.P.; Woolcock, B.W.; Rui, L.; Kawahara, M.; Farinha, P.; Johnson, N.A.; Zhao, Y.; Telenius, A.; Neriah, S.B.; et al. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature 2011, 471, 377–381. [Google Scholar] [CrossRef] [Green Version]
  118. Steidl, C.; Woolcock, B.; Rogic, S.; Ben-Neriah, S.; Telenius, A.; Drake, M.; Siebert, R.; Gascoyne, R.D. Inactivating Gene Alterations of MHC Class II Transactivator CIITA Are Recurrent in Primary Mediastinal B Cell Lymphoma and Hodgkin Lymphoma. Blood 2011, 118, 437. [Google Scholar] [CrossRef]
  119. Yazawa, T.; Ito, T.; Kamma, H.; Suzuki, T.; Okudela, K.; Hayashi, H.; Horiguchi, H.; Ogata, T.; Mitsui, H.; Ikeda, M.; et al. Complicated mechanisms of class II transactivator transcription deficiency in small cell lung cancer and neuroblastoma. Am. J. Pathol. 2002, 161, 291–300. [Google Scholar] [CrossRef] [Green Version]
  120. Holling, T.M.; Bergevoet, M.W.; Wilson, L.; Van Eggermond, M.C.; Schooten, E.; Steenbergen, R.D.; Snijders, P.J.; Jager, M.J.; Van den Elsen, P.J. A role for EZH2 in silencing of IFN-gamma inducible MHC2TA transcription in uveal melanoma. J. Immunol. 2007, 179, 5317–5325. [Google Scholar] [CrossRef] [Green Version]
  121. Martin, B.K.; Frelinger, J.G.; Ting, J.P. Combination gene therapy with CD86 and the MHC class II transactivator in the control of lung tumor growth. J. Immunol. 1999, 162, 6663–6670. [Google Scholar] [CrossRef] [PubMed]
  122. Bou Nasser Eddine, F.; Forlani, G.; Lombardo, L.; Tedeschi, A.; Tosi, G.; Accolla, R.S. CIITA-driven MHC class II expressing tumor cells can efficiently prime naive CD4 + TH cells in vivo and vaccinate the host against parental MHC-II-negative tumor cells. Oncoimmunology 2017, 6, e1261777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Frangione, V.; Mortara, L.; Castellani, P.; De Lerma Barbaro, A.; Accolla, R.S. CIITA-driven MHC-II positive tumor cells: Preventive vaccines and superior generators of antitumor CD4+ T lymphocytes for immunotherapy. Int. J. Cancer 2010, 127, 1614–1624. [Google Scholar] [CrossRef] [PubMed]
  124. Pulaski, B.A.; Ostrand-Rosenberg, S. Reduction of established spontaneous mammary carcinoma metastases following immunotherapy with major histocompatibility complex class II and B7.1 cell-based tumor vaccines. Cancer Res. 1998, 58, 1486–1493. [Google Scholar]
  125. Guillerey, C.; Huntington, N.D.; Smyth, M.J. Targeting Natural Killer Cells in Cancer Immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef]
  126. Tseng, H.C.; Bui, V.; Man, Y.G.; Cacalano, N.; Jewett, A. Induction of Split Anergy Conditions Natural Killer Cells to Promote Differentiation of Stem Cells Through Cell-Cell Contact and Secreted Factors. Front. Immunol. 2014, 5, 269. [Google Scholar] [CrossRef]
  127. Kaur, K.; Topchyan, P.; Kozlowska, A.K.; Ohanian, N.; Chiang, J.; Maung, P.O.; Park, S.H.; Ko, M.W.; Fang, C.; Nishimura, I.; et al. Super-Charged NK Cells Inhibit Growth and Progression of Stem-Like/Poorly Differentiated Oral Tumors In Vivo in Humanized BLT Mice; Effect on Tumor Differentiation and Response to Chemotherapeutic Drugs. Oncoimmunology 2018, 7, e1426518. [Google Scholar] [CrossRef] [Green Version]
  128. Ruscetti, M.; Leibold, J.; Bott, M.J.; Fennell, M.; Kulick, A.; Salgado, N.R.; Chen, C.C.; Ho, Y.J.; Sanchez-Rivera, F.J.; Feucht, J.; et al. NK Cell-Mediated Cytotoxicity Contributes to Tumor Control by a Cytostatic Drug Combination. Science 2018, 362, 1416–1422. [Google Scholar] [CrossRef] [Green Version]
  129. Ferlazzo, G.; Morandi, B. Cross-Talks Between Natural Killer Cells and Distinct Subsets of Dendritic Cells. Front. Immunol. 2014, 5, 159. [Google Scholar] [CrossRef]
  130. Walzer, T.; Dalod, M.; Robbins, S.H.; Zitvogel, L.; Vivier, E. Natural-Killer Cells and Dendritic Cells: “L’union Fait La Force”. Blood 2005, 106, 2252–2258. [Google Scholar] [CrossRef] [Green Version]
  131. Moretta, L.; Ferlazzo, G.; Bottino, C.; Vitale, M.; Pende, D.; Mingari, M.C.; Moretta, A. Effector and Regulatory Events During Natural Killer-Dendritic Cell Interactions. Immunol. Rev. 2006, 214, 219–228. [Google Scholar] [CrossRef] [PubMed]
  132. Kalinski, P.; Mailliard, R.B.; Giermasz, A.; Zeh, H.J.; Basse, P.; Bartlett, D.L.; Kirkwood, J.M.; Lotze, M.T.; Herberman, R.B. Natural Killer-Dendritic Cell Cross-Talk in Cancer Immunotherapy. Expert Opin. Biol. Ther. 2005, 5, 1303–1315. [Google Scholar] [CrossRef] [PubMed]
  133. Lee, S.C.; Srivastava, R.M.; Lopez-Albaitero, A.; Ferrone, S.; Ferris, R.L. Natural Killer (NK): Dendritic Cell (DC) Cross Talk Induced by Therapeutic Monoclonal Antibody Triggers Tumor Antigen-Specific T Cell Immunity. Immunol. Res. 2011, 50, 248–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Salmon, H.; Idoyaga, J.; Rahman, A.; Leboeuf, M.; Remark, R.; Jordan, S.; Casanova-Acebes, M.; Khudoynazarova, M.; Agudo, J.; Tung, N.; et al. Expansion and Activation of CD103(+) Dendritic Cell Progenitors At the Tumor Site Enhances Tumor Responses to Therapeutic Pd-L1 and BRAF Inhibition. Immunity 2016, 44, 924–938. [Google Scholar] [CrossRef] [Green Version]
  135. Spranger, S.; Dai, D.; Horton, B.; Gajewski, T.F. Tumor-Residing Batf3 Dendritic Cells are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017, 31, 711–723:e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Oba, T.; Long, M.D.; Keler, T.; Marsh, H.C.; Minderman, H.; Abrams, S.I.; Liu, S.; Ito, F. Overcoming Primary and Acquired Resistance to anti-PD-L1 Therapy by Induction and Activation of Tumor-Residing Cdc1s. Nat. Commun. 2020, 11, 5415. [Google Scholar] [CrossRef]
  137. Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 Into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e14. [Google Scholar] [CrossRef] [Green Version]
  138. Barry, K.C.; Hsu, J.; Broz, M.L.; Cueto, F.J.; Binnewies, M.; Combes, A.J.; Nelson, A.E.; Loo, K.; Kumar, R.; Rosenblum, M.D.; et al. A Natural Killer-Dendritic Cell Axis Defines Checkpoint Therapy-Responsive Tumor Microenvironments. Nat. Med. 2018, 24, 1178–1191. [Google Scholar] [CrossRef]
  139. Fujihara, A.; Kurooka, M.; Miki, T.; Kaneda, Y. Intratumoral Injection of Inactivated Sendai Virus Particles Elicits Strong Antitumor Activity by Enhancing Local CXCL10 Expression and Systemic NK Cell Activation. Cancer Immunol. Immunother. 2008, 57, 73–84. [Google Scholar] [CrossRef]
  140. Kim, N.; Kim, H.S. Targeting Checkpoint Receptors and Molecules for Therapeutic Modulation of Natural Killer Cells. Front. Immunol. 2018, 9, 2041. [Google Scholar] [CrossRef]
  141. Klose, C.; Berchtold, S.; Schmidt, M.; Beil, J.; Smirnow, I.; Venturelli, S.; Burkard, M.; Handgretinger, R.; Lauer, U.M. Biological Treatment of Pediatric Sarcomas by Combined Virotherapy and NK Cell Therapy. BMC Cancer 2019, 19, 1172. [Google Scholar] [CrossRef] [Green Version]
  142. Imai, K.; Matsuyama, S.; Miyake, S.; Suga, K.; Nakachi, K. Natural Cytotoxic Activity of Peripheral-Blood Lymphocytes and Cancer Incidence: An 11-Year Follow-Up Study of a General Population. Lancet 2000, 356, 1795–1799. [Google Scholar] [CrossRef] [PubMed]
  143. Balch, C.M.; Tilden, A.B.; Dougherty, P.A.; Cloud, G.A. Depressed Levels of Granular Lymphocytes With Natural Killer (NK) Cell Function in 247 Cancer Patients. Ann. Surg. 1983, 198, 192–199. [Google Scholar] [CrossRef] [PubMed]
  144. Angka, L.; Khan, S.T.; Kilgour, M.K.; Xu, R.; Kennedy, M.A.; Auer, R.C. Dysfunctional Natural Killer Cells in the Aftermath of Cancer Surgery. Int. J. Mol. Sci. 2017, 18, 1787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Cong, J.; Wang, X.; Zheng, X.; Wang, D.; Fu, B.; Sun, R.; Tian, Z.; Wei, H. Dysfunction of Natural Killer Cells by FBP1-Induced Inhibition of Glycolysis During Lung Cancer Progression. Cell Metab. 2018, 28, 243–255.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Shen, M.J.; Xu, L.J.; Yang, L.; Tsai, Y.; Keng, P.C.; Chen, Y.; Lee, S.O.; Chen, Y. Radiation Alters PDL1/NKG2D Ligand Levels in Lung Cancer Cells and Leads to Immune Escape From NK Cell Cytotoxicity Via IL-6-MEK/Erk Signaling Pathway. Oncotarget 2017, 8, 80506–80520. [Google Scholar] [CrossRef] [Green Version]
  147. Mantovani, S.; Oliviero, B.; Lombardi, A.; Varchetta, S.; Mele, D.; Sangiovanni, A.; Rossi, G.; Donadon, M.; Torzilli, G.; Soldani, C.; et al. Deficient Natural Killer Cell Nkp30-Mediated Function and Altered Ncr3 Splice Variants in Hepatocellular Carcinoma. Hepatology 2019, 69, 1165–1179. [Google Scholar] [CrossRef] [PubMed]
  148. Zheng, Y.; Li, Y.; Lian, J.; Yang, H.; Li, F.; Zhao, S.; Qi, Y.; Zhang, Y.; Huang, L. TNF-α-Induced Tim-3 Expression Marks the Dysfunction of Infiltrating Natural Killer Cells in Human Esophageal Cancer. J. Trans. Med. 2019, 17, 165. [Google Scholar] [CrossRef] [Green Version]
  149. Ciurea, S.O.; Schafer, J.R.; Bassett, R.; Denman, C.J.; Cao, K.; Willis, D.; Rondon, G.; Chen, J.; Soebbing, D.; Kaur, I.; et al. Phase 1 Clinical Trial Using mbIL21 Ex Vivo-Expanded Donor-Derived NK Cells After Haploidentical Transplantation. Blood 2017, 130, 1857–1868. [Google Scholar] [CrossRef] [Green Version]
  150. Baggio, L.; Laureano, Á.M.; Silla, L.; Lee, D.A. Natural Killer Cell Adoptive Immunotherapy: Coming of Age. Clin. Immunol. 2017, 177, 3–11. [Google Scholar] [CrossRef]
  151. Pistoia, V.; Tumino, N.; Vacca, P.; Veneziani, I.; Moretta, A.; Locatelli, F.; Moretta, L. Human gammadelta T-Cells: From Surface Receptors to the Therapy of High-Risk Leukemias. Front. Immunol. 2018, 9, 984. [Google Scholar] [CrossRef] [Green Version]
  152. Minculescu, L.; Sengelov, H. The role of gamma delta T cells in haematopoietic stem cell transplantation. Scand. J. Immunol. 2015, 81, 459–468. [Google Scholar] [CrossRef]
  153. Handgretinger, R.; Schilbach, K. The potential role of gammadelta T cells after allogeneic HCT for leukemia. Blood 2018, 131, 1063–1072. [Google Scholar] [CrossRef] [Green Version]
  154. Parker, C.M.; Groh, V.; Band, H.; Porcelli, S.A.; Morita, C.; Fabbi, M.; Glass, D.; Strominger, J.L.; Brenner, M.B. Evidence for extrathymic changes in the T cell receptor gamma/delta repertoire. J. Exp. Med. 1990, 171, 1597–1612. [Google Scholar] [CrossRef] [PubMed]
  155. Kunzmann, V.; Wilhelm, M. Anti-lymphoma effect of gammadelta T cells. Leuk. Lymphoma 2005, 46, 671–680. [Google Scholar] [CrossRef] [PubMed]
  156. Di Lorenzo, B.; Simoes, A.E.; Caiado, F.; Tieppo, P.; Correia, D.V.; Carvalho, T.; da Silva, M.G.; Dechanet-Merville, J.; Schumacher, T.N.; Prinz, I.; et al. Broad Cytotoxic Targeting of Acute Myeloid Leukemia by Polyclonal Delta One T Cells. Cancer Immunol. Res. 2019, 7, 552–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Kabelitz, D.; He, W. The multifunctionality of human Vgamma9Vdelta2 gammadelta T cells: Clonal plasticity or distinct subsets? Scand. J. Immunol. 2012, 76, 213–222. [Google Scholar] [CrossRef] [PubMed]
  158. Gentles, A.J.; Newman, A.M.; Liu, C.L.; Bratman, S.V.; Feng, W.; Kim, D.; Nair, V.S.; Xu, Y.; Khuong, A.; Hoang, C.D.; et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 2015, 21, 938–945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Silva-Santos, B.; Mensurado, S.; Coffelt, S.B. gammadelta T cells: Pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer 2019, 19, 392–404. [Google Scholar] [CrossRef] [Green Version]
  160. Sandberg, Y.; Almeida, J.; Gonzalez, M.; Lima, M.; Barcena, P.; Szczepanski, T.; van Gastel-Mol, E.J.; Wind, H.; Balanzategui, A.; van Dongen, J.J.; et al. TCRgammadelta+ large granular lymphocyte leukemias reflect the spectrum of normal antigen-selected TCRgammadelta+ T-cells. Leukemia 2006, 20, 505–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Qi, C.; Wang, Y.; Li, P.; Zhao, J. Gamma Delta T Cells and Their Pathogenic Role in Psoriasis. Front. Immunol. 2021, 12, 627139. [Google Scholar] [CrossRef] [PubMed]
  162. Kenna, T.; Golden-Mason, L.; Norris, S.; Hegarty, J.E.; O’Farrelly, C.; Doherty, D.G. Distinct subpopulations of gamma delta T cells are present in normal and tumor-bearing human liver. Clin. Immunol. 2004, 113, 56–63. [Google Scholar] [CrossRef] [PubMed]
  163. Davey, M.S.; Willcox, C.R.; Hunter, S.; Kasatskaya, S.A.; Remmerswaal, E.B.M.; Salim, M.; Mohammed, F.; Bemelman, F.J.; Chudakov, D.M.; Oo, Y.H.; et al. The human Vdelta2+ T-cell compartment comprises distinct innate-like Vgamma9+ and adaptive Vgamma9− subsets. Nat. Commun. 2018, 9, 1760. [Google Scholar] [CrossRef] [PubMed]
  164. Rigau, M.; Ostrouska, S.; Fulford, T.S.; Johnson, D.N.; Woods, K.; Ruan, Z.; McWilliam, H.E.G.; Hudson, C.; Tutuka, C.; Wheatley, A.K.; et al. Butyrophilin 2A1 is essential for phosphoantigen reactivity by gammadelta T cells. Science 2020, 367, eaay5516. [Google Scholar] [CrossRef] [PubMed]
  165. Guerra, B.; Recio, C.; Aranda-Tavio, H.; Guerra-Rodriguez, M.; Garcia-Castellano, J.M.; Fernandez-Perez, L. The Mevalonate Pathway, a Metabolic Target in Cancer Therapy. Front. Oncol. 2021, 11, 626971. [Google Scholar] [CrossRef]
  166. Freed-Pastor, W.A.; Mizuno, H.; Zhao, X.; Langerod, A.; Moon, S.H.; Rodriguez-Barrueco, R.; Barsotti, A.; Chicas, A.; Li, W.; Polotskaia, A.; et al. Mutant p53 disrupts mammary tissue architecture via the mevalonate pathway. Cell 2012, 148, 244–258. [Google Scholar] [CrossRef] [Green Version]
  167. Luoma, A.M.; Castro, C.D.; Adams, E.J. gammadelta T cell surveillance via CD1 molecules. Trends Immunol. 2014, 35, 613–621. [Google Scholar] [CrossRef]
  168. Bai, L.; Picard, D.; Anderson, B.; Chaudhary, V.; Luoma, A.; Jabri, B.; Adams, E.J.; Savage, P.B.; Bendelac, A. The majority of CD1d-sulfatide-specific T cells in human blood use a semiinvariant Vdelta1 TCR. Eur. J. Immunol. 2012, 42, 2505–2510. [Google Scholar] [CrossRef] [Green Version]
  169. Russano, A.M.; Agea, E.; Corazzi, L.; Postle, A.D.; De Libero, G.; Porcelli, S.; de Benedictis, F.M.; Spinozzi, F. Recognition of pollen-derived phosphatidyl-ethanolamine by human CD1d-restricted gamma delta T cells. J. Allergy Clin. Immunol. 2006, 117, 1178–1184. [Google Scholar] [CrossRef]
  170. Agea, E.; Russano, A.; Bistoni, O.; Mannucci, R.; Nicoletti, I.; Corazzi, L.; Postle, A.D.; De Libero, G.; Porcelli, S.A.; Spinozzi, F. Human CD1-restricted T cell recognition of lipids from pollens. J. Exp. Med. 2005, 202, 295–308. [Google Scholar] [CrossRef]
  171. Mangan, B.A.; Dunne, M.R.; O’Reilly, V.P.; Dunne, P.J.; Exley, M.A.; O’Shea, D.; Scotet, E.; Hogan, A.E.; Doherty, D.G. Cutting edge: CD1d restriction and Th1/Th2/Th17 cytokine secretion by human Vdelta3 T cells. J. Immunol. 2013, 191, 30–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Liang, F.; Zhang, C.; Guo, H.; Gao, S.H.; Yang, F.Y.; Zhou, G.B.; Wang, G.Z. Comprehensive analysis of BTN3A1 in cancers: Mining of omics data and validation in patient samples and cellular models. FEBS Open Bio 2021, 11, 2586–2599. [Google Scholar] [CrossRef] [PubMed]
  173. Nair, S.; Dhodapkar, M.V. Natural Killer T Cells in Cancer Immunotherapy. Front. Immunol. 2017, 8, 1178. [Google Scholar] [CrossRef] [Green Version]
  174. Bauer, S.; Groh, V.; Wu, J.; Steinle, A.; Phillips, J.H.; Lanier, L.L.; Spies, T. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999, 285, 727–729. [Google Scholar] [CrossRef]
  175. Simoes, A.E.; Di Lorenzo, B.; Silva-Santos, B. Molecular Determinants of Target Cell Recognition by Human gammadelta T Cells. Front. Immunol. 2018, 9, 929. [Google Scholar] [CrossRef] [Green Version]
  176. Groh, V.; Rhinehart, R.; Secrist, H.; Bauer, S.; Grabstein, K.H.; Spies, T. Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc. Natl. Acad. Sci. USA 1999, 96, 6879–6884. [Google Scholar] [CrossRef]
  177. Kong, Y.; Cao, W.; Xi, X.; Ma, C.; Cui, L.; He, W. The NKG2D ligand ULBP4 binds to TCRgamma9/delta2 and induces cytotoxicity to tumor cells through both TCRgammadelta and NKG2D. Blood 2009, 114, 310–317. [Google Scholar] [CrossRef] [PubMed]
  178. Lanca, T.; Correia, D.V.; Moita, C.F.; Raquel, H.; Neves-Costa, A.; Ferreira, C.; Ramalho, J.S.; Barata, J.T.; Moita, L.F.; Gomes, A.Q.; et al. The MHC class Ib protein ULBP1 is a nonredundant determinant of leukemia/lymphoma susceptibility to gammadelta T-cell cytotoxicity. Blood 2010, 115, 2407–2411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Kabelitz, D.; Serrano, R.; Kouakanou, L.; Peters, C.; Kalyan, S. Cancer immunotherapy with gammadelta T cells: Many paths ahead of us. Cell Mol. Immunol. 2020, 17, 925–939. [Google Scholar] [CrossRef]
  180. Zhao, Y.; Niu, C.; Cui, J. Gamma-delta (gammadelta) T cells: Friend or foe in cancer development? J. Transl. Med. 2018, 16, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Todaro, M.; D’Asaro, M.; Caccamo, N.; Iovino, F.; Francipane, M.G.; Meraviglia, S.; Orlando, V.; La Mendola, C.; Gulotta, G.; Salerno, A.; et al. Efficient killing of human colon cancer stem cells by gammadelta T lymphocytes. J. Immunol. 2009, 182, 7287–7296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Dieli, F.; Vermijlen, D.; Fulfaro, F.; Caccamo, N.; Meraviglia, S.; Cicero, G.; Roberts, A.; Buccheri, S.; D’Asaro, M.; Gebbia, N.; et al. Targeting human γδ T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res. 2007, 67, 7450–7457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Couzi, L.; Pitard, V.; Sicard, X.; Garrigue, I.; Hawchar, O.; Merville, P.; Moreau, J.F.; Dechanet-Merville, J. Antibody-dependent anti-cytomegalovirus activity of human gammadelta T cells expressing CD16 (FcgammaRIIIa). Blood 2012, 119, 1418–1427. [Google Scholar] [CrossRef] [PubMed]
  184. Tokuyama, H.; Hagi, T.; Mattarollo, S.R.; Morley, J.; Wang, Q.; So, H.F.; Moriyasu, F.; Nieda, M.; Nicol, A.J. V gamma 9 V delta 2 T cell cytotoxicity against tumor cells is enhanced by monoclonal antibody drugs–rituximab and trastuzumab. Int. J. Cancer 2008, 122, 2526–2534. [Google Scholar] [CrossRef]
  185. Dar, A.A.; Patil, R.S.; Chiplunkar, S.V. Insights into the Relationship between Toll Like Receptors and Gamma Delta T Cell Responses. Front. Immunol. 2014, 5, 366. [Google Scholar] [CrossRef] [Green Version]
  186. O’Neill, L.A.; Golenbock, D.; Bowie, A.G. The History of Toll-like Receptors—Redefining Innate Immunity. Nat. Rev. Immunol. 2013, 13, 453–460. [Google Scholar] [CrossRef]
  187. Corsale, A.M.; Di Simone, M.; Lo Presti, E.; Picone, C.; Dieli, F.; Meraviglia, S. Metabolic Changes in Tumor Microenvironment: How Could They Affect gammadelta T Cells Functions? Cells 2021, 10, 2896. [Google Scholar] [CrossRef]
  188. Fowler, D.W.; Copier, J.; Dalgleish, A.G.; Bodman-Smith, M.D. Zoledronic acid renders human M1 and M2 macrophages susceptible to Vdelta2+ gammadelta T cell cytotoxicity in a perforin-dependent manner. Cancer Immunol. Immunother. 2017, 66, 1205–1215. [Google Scholar] [CrossRef] [Green Version]
  189. Lin, Y.; Xu, J.; Lan, H. Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J. Hematol. Oncol. 2019, 12, 76. [Google Scholar] [CrossRef]
  190. Brandes, M.; Willimann, K.; Moser, B. Professional antigen-presentation function by human gammadelta T Cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef]
  191. Brandes, M.; Willimann, K.; Bioley, G.; Levy, N.; Eberl, M.; Luo, M.; Tampe, R.; Levy, F.; Romero, P.; Moser, B. Cross-presenting human gammadelta T cells induce robust CD8+ alphabeta T cell responses. Proc. Natl. Acad. Sci. USA 2009, 106, 2307–2312. [Google Scholar] [CrossRef] [PubMed]
  192. Muto, M.; Baghdadi, M.; Maekawa, R.; Wada, H.; Seino, K. Myeloid molecular characteristics of human gammadelta T cells support their acquisition of tumor antigen-presenting capacity. Cancer Immunol. Immunother. 2015, 64, 941–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Scotet, E.; Nedellec, S.; Devilder, M.C.; Allain, S.; Bonneville, M. Bridging innate and adaptive immunity through gammadelta T-dendritic cell crosstalk. Front. Biosci. 2008, 13, 6872–6885. [Google Scholar] [CrossRef] [Green Version]
  194. van Beek, J.J.; Wimmers, F.; Hato, S.V.; de Vries, I.J.; Skold, A.E. Dendritic cell cross talk with innate and innate-like effector cells in antitumor immunity: Implications for DC vaccination. Crit. Rev. Immunol. 2014, 34, 517–536. [Google Scholar] [CrossRef] [PubMed]
  195. Maniar, A.; Zhang, X.; Lin, W.; Gastman, B.R.; Pauza, C.D.; Strome, S.E.; Chapoval, A.I. Human gammadelta T lymphocytes induce robust NK cell-mediated antitumor cytotoxicity through CD137 engagement. Blood 2010, 116, 1726–1733. [Google Scholar] [CrossRef] [Green Version]
  196. Ladel, C.H.; Blum, C.; Kaufmann, S.H. Control of natural killer cell-mediated innate resistance against the intracellular pathogen Listeria monocytogenes by gamma/delta T lymphocytes. Infect. Immun. 1996, 64, 1744–1749. [Google Scholar] [CrossRef] [Green Version]
  197. Jacobsen, E.A.; Jackson, D.J.; Heffler, E.; Mathur, S.K.; Bredenoord, A.J.; Pavord, I.D.; Akuthota, P.; Roufosse, F.; Rothenberg, M.E. Eosinophil knockout humans: Uncovering the role of eosinophils through eosinophil-directed biological therapies. Annu. Rev. Immunol. 2021, 39, 719–757. [Google Scholar] [CrossRef]
  198. Reinbach, G. Ueber das Verhalten der Leukocyten bei malignen Tumoren. Arch. Klin. Chir. 1893, 46, 486–562. [Google Scholar]
  199. Grisaru-Tal, S.; Itan, M.; Grass, D.G.; Torres-Roca, J.; Eschrich, S.A.; Gordon, Y.; Dolitzky, A.; Hazut, I.; Avlas, S.; Jacobsen, E.A.; et al. Primary tumors from mucosal barrier organs drive unique eosinophil infiltration patterns and clinical associations. Oncoimmunology 2020, 10, 1859732. [Google Scholar] [CrossRef]
  200. Grisaru-Tal, S.; Itan, M.; Klion, A.D.; Munitz, A. A new dawn for eosinophils in the tumour microenvironment. Nat. Rev. Cancer 2020, 20, 594–607. [Google Scholar] [CrossRef]
  201. Rodriguez-Rodriguez, N.; Gogoi, M.; McKenzie, A.N.J. Group 2 innate lymphoid cells: Team players in regulating asthma. Annu. Rev. Immunol. 2021, 39, 167–198. [Google Scholar] [CrossRef]
  202. Nussbaum, J.C.; Van Dyken, S.J.; von Moltke, J.; Cheng, L.E.; Mohapatra, A.; Molofsky, A.B.; Thornton, E.E.; Krummel, M.F.; Chawla, A.; Liang, H.E.; et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 2013, 502, 245–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Maggi, E.; Veneziani, I.; Moretta, L.; Cosmi, L.; Annunziato, F. Group 2 innate lymphoid cells: A double-edged sword in cancer? Cancers 2020, 12, 3452. [Google Scholar] [CrossRef] [PubMed]
  204. Jacquelot, N.; Seillet, C.; Wang, M.; Pizzolla, A.; Liao, Y.; Hediyeh-Zadeh, S.; Grisaru-Tal, S.; Louis, C.; Huang, Q.; Schreuder, J.; et al. Blockade of the co-inhibitory molecule PD-1 unleashes ILC2-dependent antitumor immunity in melanoma. Nat. Immunol. 2021, 22, 851–864. [Google Scholar] [CrossRef]
  205. Ikutani, M.; Yanagibashi, T.; Ogasawara, M.; Tsuneyama, K.; Yamamoto, S.; Hattori, Y.; Kouro, T.; Itakura, A.; Nagai, Y.; Takaki, S.; et al. Identification of innate IL-5-producing cells and their role in lung eosinophil regulation and antitumor immunity. J. Immunol. 2012, 188, 703–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Martin, N.T.; Martin, M.U. Interleukin 33 is a guardian of barriers and a local alarmin. Nat. Immunol. 2016, 17, 122–131. [Google Scholar] [CrossRef]
  207. Lucarini, V.; Ziccheddu, G.; Macchia, I.; La Sorsa, V.; Peschiaroli, F.; Buccione, C.; Sistigu, A.; Sanchez, M.; Andreone, S.; D’Urso, M.T.; et al. IL-33 restricts tumor growth and inhibits pulmonary metastasis in melanoma-bearing mice through eosinophils. Oncoimmunology 2017, 6, e1317420. [Google Scholar] [CrossRef] [Green Version]
  208. Gao, K.; Li, X.; Zhang, L.; Bai, L.; Dong, W.; Gao, K.; Shi, G.; Xia, X.; Wu, L.; Zhang, L. Transgenic expression of IL-33 activates CD8+ T cells and NK cells and inhibits tumor growth and metastasis in mice. Cancer Lett. 2013, 335, 463–471. [Google Scholar] [CrossRef]
  209. Munitz, A.; Bachelet, I.; Fraenkel, S.; Katz, G.; Mandelboim, O.; Simon, H.U.; Moretta, L.; Colonna, M.; Levi-Schaffer, F. 2B4 (CD244) is expressed and functional on human eosinophils. J. Immunol. 2005, 174, 110–118. [Google Scholar] [CrossRef] [Green Version]
  210. Munitz, A.; Bachelet, I.; Eliashar, R.; Moretta, A.; Moretta, L.; Levi-Schaffer, F. The inhibitory receptor IRp60 (CD300a) suppresses the effects of IL-5, GM-CSF and eotaxin on human peripheral blood eosinophils. Blood 2006, 107, 1996–2003. [Google Scholar] [CrossRef] [Green Version]
  211. Pesce, S.; Thoren, F.B.; Cantoni, C.; Prato, C.; Moretta, L.; Moretta, A.; Marcenaro, E. The innate immune cross-talk between NK cells and eosinophils is regulated by the interaction of natural cytotoxicity receptors with eosinophil surface ligands. Front. Immunol. 2017, 8, 510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Qi, L.; Zhang, Q.; Miao, Y.; Kang, W.; Tian, Z.; Xu, D.; Xiao, W.; Fang, F. Interleukin-33 activates and recruits natural killer cells to inhibit pulmonary metastatic cancer development. Int. J. Cancer 2020, 146, 1421–1434. [Google Scholar] [CrossRef] [PubMed]
  213. Schuijs, M.J.; Png, S.; Richard, A.C.; Tsyben, A.; Hamm, G.; Stockis, J.; Garcia, C.; Pinaud, S.; Nicholls, A.; Ros, X.R.; et al. ILC2-driven innate immune checkpoint mechanism antagonizes NK cell antimetastatic function in the lung. Nat. Immunol. 2020, 21, 998–1009. [Google Scholar] [CrossRef]
  214. Grisaru-Tal, S.; Dulberg, S.; Beck, L.; Zhang, C.; Itan, M.; Hediyeh-Zadeh, S.; Caldwell, J.; Rozenberg, P.; Dolitzky, A.; Avlas, S.; et al. Metastasis-entrained eosinophils enhance lymphocytemediated antitumor immunity. Cancer Res. 2021, 81, 5555–5571. [Google Scholar] [CrossRef]
  215. Reichman, H.; Itan, M.; Rozenberg, P.; Yarmolovski, T.; Brazowski, E.; Varol, C.; Gluck, N.; Shapira, S.; Arber, N.; Qimron, U.H.; et al. Activated eosinophils exert antitumorigenic activities in colorectal cancer. Cancer Immunol. Res. 2019, 7, 388–400. [Google Scholar] [CrossRef] [Green Version]
  216. Dolitzky, A.; Shapira, G.; Grisaru-Tal, S.; Hazut, I.; Avlas, S.; Gordon, Y.; Itan, M.; Shomron, N.; Munitz, A. Transcriptional profiling of mouse eosinophils identifies distinct gene signatures following cellular activation. Front. Immunol. 2021, 12, 802839. [Google Scholar] [CrossRef]
  217. Carretero, R.; Sektioglu, I.M.; Garbi, N.; Salgado, O.C.; Beckhove, P.; Hämmerling, G.J. Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8+ T cells. Nat. Immunol. 2015, 16, 609–617. [Google Scholar] [CrossRef]
  218. Akuthota, P.; Wang, H.B.; Spencer, L.A.; Weller, P.F. Immunoregulatory roles of eosinophils: A new look at a familiar cell. Clin. Exp. Allergy 2008, 38, 1254–1263. [Google Scholar] [CrossRef]
  219. Simon, H.U.; Plötz, S.; Simon, D.; Seitzer, U.; Braathen, L.R.; Menz, G.; Straumann, A.; Dummer, R.; Levi-Schaffer, F. Interleukin-2 primes eosinophil degranulation in hypereosinophilia and Wells’ syndrome. Eur. J. Immunol. 2003, 33, 834–839. [Google Scholar] [CrossRef] [PubMed]
  220. Sosman, J.A.; Bartemes, K.; Offord, K.P.; Kita, H.; Fisher, S.G.; Kefer, C.; Ellis, T.A.; Fisher, R.I.; Higgins, T.J.; Gleich, G.J. Evidence for eosinophil activation in cancer patients receiving recombinant interleukin-4: Effects of interleukin-4 alone and following interleukin-2 administration. Clin. Cancer Res. 1995, 1, 805–812. [Google Scholar] [PubMed]
  221. Ellem, K.A.; O’Rourke, M.G.; Johnson, G.R.; Parry, G.; Misko, I.S.; Schmidt, C.W.; Parsons, P.G.; Burrows, S.R.; Cross, S.; Fell, A.; et al. A case report: Immune responses and clinical course of the first human use of granulocyte/macrophage-colony- stimulating-factor-transduced autologous melanoma cells for immunotherapy. Cancer Immunol. Immunother. 1997, 44, 10–20. [Google Scholar] [CrossRef] [PubMed]
  222. Gebhardt, C.; Sevko, A.; Jiang, H.; Lichtenberger, R.; Reith, M.; Tarnanidis, K.; Holland-Letz, T.; Umansky, L.; Beckhove, P.; Sucker, A.; et al. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin. Cancer Res. 2015, 21, 5453–5459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Moreira, A.; Leisgang, W.; Schuler, G.; Heinzerling, L. Eosinophilic count as a biomarker for prognosis of melanoma patients and its importance in the response to immunotherapy. Immunotherapy 2017, 9, 115–121. [Google Scholar] [CrossRef]
  224. Weide, B.; Martens, A.; Hassel, J.C.; Berking, C.; Postow, M.A.; Bisschop, K.; Simeone, E.; Mangana, J.; Schilling, B.; Di Giacomo, A.M.; et al. Baseline biomarkers for outcome of melanoma patients treated with pembrolizumab. Clin. Cancer Res. 2016, 22, 5487–5496. [Google Scholar] [CrossRef] [Green Version]
  225. Alves, A.; Dias, M.; Campainha, S.; Barroso, A. Peripheral blood eosinophilia may be a prognostic biomarker in non-small cell lung cancer patients treated with immunotherapy. J. Thorac. Dis. 2021, 13, 2716–2727. [Google Scholar] [CrossRef]
  226. Herrmann, T.; Ginzac, A.; Molnar, I.; Bailly, S.; Durando, X.; Mahammedi, H. Eosinophil counts as a relevant prognostic marker for response to nivolumab in the management of renal cell carcinoma: A retrospective study. Cancer Med. 2021, 10, 6705–6713. [Google Scholar] [CrossRef] [PubMed]
  227. Ghebeh, H.; Elshenawy, M.A.; AlSayed, A.D.; Al-Tweigeri, T. Peripheral blood eosinophil count is associated with response to chemoimmunotherapy in metastatic triple-negative breast cancer. Immunotherapy 2022, 14, 189–199. [Google Scholar] [CrossRef]
  228. Nishikawa, D.; Suzuki, H.; Beppu, S.; Terada, H.; Sawabe, M.; Kadowaki, S.; Sone, M.; Hanai, N. Eosinophil prognostic scores for patients with head and neck squamous cell carcinoma treated with nivolumab. Cancer Sci. 2021, 112, 339–346. [Google Scholar] [CrossRef]
  229. Lai, W.; Xie, H.; Liu, Y.; Zheng, F.; Zhang, Y.; Lei, Q.; Lv, L.; Dong, J.; Song, J.; Gao, X.; et al. Human pluripotent stem cell-derived eosinophils reveal potent cytotoxicity against solid tumors. Stem Cell Rep. 2021, 16, 1697–1704. [Google Scholar] [CrossRef]
  230. Lynch, J.B.; Hsiao, E.Y. Microbiomes as sources of emergent host phenotypes. Science 2019, 365, 1405–1409. [Google Scholar] [CrossRef]
  231. Amieva, M.; Peek, R.M. Pathobiology of Helicobacter pylori–induced gastric cancer. Gastroenterology 2016, 150, 64–78. [Google Scholar] [CrossRef] [Green Version]
  232. Moss, S.F. The clinical evidence linking Helicobacter pylori to gastric cancer. Cell Mol. Gastroenterol. Hepatol. 2017, 3, 183–191. [Google Scholar] [CrossRef] [Green Version]
  233. Flanagan, L.; Schmid, J.; Ebert, M.; Soucek, P.; Kunicka, T.; Liska, V.; Bruha, J.; Neary, P.; Dezeeuw, N.; Tommasino, M.; et al. Fusobacterium nucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome. Eur. J. Clin. Microbiol. Infect Dis. 2014, 33, 1381–1390. [Google Scholar] [CrossRef]
  234. Geller, L.T.; Barzily-Rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 357, 1156–1160. [Google Scholar] [CrossRef] [Green Version]
  235. Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef] [Green Version]
  236. Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; San Lucas, A.; et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 2019, 178, 795–806.e12. [Google Scholar] [CrossRef] [PubMed]
  237. Ma, C.; Han, M.; Heinrich, B.; Fu, Q.; Zhang, Q.; Sandhu, M.; Agdashian, D.; Terabe, M.; Berzofsky, J.A.; Fako, V.; et al. Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018, 360, eaan5931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Loo, T.M.; Kamachi, F.; Watanabe, Y.; Yoshimoto, S.; Kanda, H.; Arai, Y.; Nakajima-Takagi, Y.; Iwama, A.; Koga, T.; Sugimoto, Y.; et al. Gut microbiota promotes obesity-associated liver cancer through PGE2-mediated suppression of antitumor immunity. Cancer Discov. 2017, 7, 522–538. [Google Scholar] [CrossRef] [Green Version]
  239. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen–gut microbiome axis: Physiological and clinical implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef] [Green Version]
  240. Flores, R.; Shi, J.; Fuhrman, B.; Xu, X.; Veenstra, T.D.; Gail, M.H.; Gajer, P.; Ravel, J.; Goedert, J.J. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: A cross-sectional study. J. Transl. Med. 2012, 10, 253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Kwa, M.; Plottel, C.S.; Blaser, M.J.; Adams, S. The intestinal microbiome and estrogen receptor-positive female breast cancer. J. Natl. Cancer Inst. 2016, 108, djw029. [Google Scholar]
  242. Rao, V.P.; Poutahidis, T.; Ge, Z.; Nambiar, P.R.; Boussahmain, C.; Wang, Y.Y.; Horwitz, B.H.; Fox, J.G.; Erdman, S.E. Innate immune inflammatory response against enteric bacteria Helicobacter hepaticus induces mammary adenocarcinoma in mice. Cancer Res. 2006, 66, 7395–7400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Eslami, S.Z.; Majidzadeh, A.K.; Halvaei, S.; Babapirali, F.; Esmaeili, R. Microbiome and breast cancer: New role for an ancient population. Front. Oncol. 2020, 10, 120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Ma, Q.; Xing, C.; Long, W.; Wang, H.Y.; Liu, Q.; Wang, R.F. Impact of microbiota on central nervous system and neurological diseases: The gut-brain axis. J. Neuroinflammation 2019, 16, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Abdul-Aziz, M.A.; Cooper, A.; Weyrich, L.S. Exploring relationships between host genome and microbiome: New insights from genome-wide association studies. Front. Microbiol. 2016, 7, 1611. [Google Scholar] [CrossRef] [Green Version]
  246. Ben-Shaanan, T.L.; Schiller, M.; Azulay-Debby, H.; Korin, B.; Boshnak, N.; Koren, T.; Krot, M.; Shakya, J.; Rahat, M.A.; Hakim, F.; et al. Modulation of anti-tumor immunity by the brain’s reward system. Nat. Commun. 2018, 9, 2723. [Google Scholar] [CrossRef] [Green Version]
  247. Xavier, J.B.; Young, V.B.; Skufca, J.; Ginty, F.; Testerman, T.; Pearson, A.T.; Macklin, P.; Mitchell, A.; Shmulevich, I.; Xie, L.; et al. The cancer microbiome: Distinguishing direct and indirect effects requires a systemic view. Trends Cancer 2020, 6, 192–204. [Google Scholar] [CrossRef] [Green Version]
  248. Fluckiger, A.; Daillère, R.; Sassi, M.; Sixt, B.S.; Liu, P.; Loos, F.; Richard, C.; Rabu, C.; Alou, M.T.; Goubet, A.G.; et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science 2020, 369, 936–942. [Google Scholar] [CrossRef]
  249. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
  250. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar]
  251. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Borgerding, J.N.; Shang, J.; Britton, G.J.; Salmon, H.; Bigenwald, C.; Maier, B.; Rose, S.R.; Mogno, I.; Kamphorst, A.O.; Merad, M.; et al. Human microbial transplant restores T cell cytotoxicity and anti-tumor response to PD-L1 blockade in gnotobiotic mice. bioRxiv 2020, 242040. [Google Scholar] [CrossRef]
  253. Mager, L.F.; Burkhard, R.; Pett, N.; Cooke, N.C.A.; Brown, K.; Ramay, H.; Paik, S.; Stagg, J.; Groves, R.A.; Gallo, M.; et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 2020, 369, 1481–1489. [Google Scholar] [CrossRef] [PubMed]
  254. Li, M.O.; Wolf, N.; Raulet, D.H.; Akkari, L.; Pittet, M.J.; Rodriguez, P.C.; Kaplan, R.N.; Munitz, A.; Zhang, Z.; Cheng, S.; et al. Innate immune cells in the tumor microenvironment. Cancer Cell 2021, 39, 725–729. [Google Scholar] [CrossRef]
Figure 1. Illustration of MHC-I deficiency and immune evasion.
Figure 1. Illustration of MHC-I deficiency and immune evasion.
Cells 12 01882 g001
Figure 2. Illustration of tsMHC-II’s role in tumor immune surveillance.
Figure 2. Illustration of tsMHC-II’s role in tumor immune surveillance.
Cells 12 01882 g002
Figure 3. Summary of NK cells’ function in anti-cancer immunity.
Figure 3. Summary of NK cells’ function in anti-cancer immunity.
Cells 12 01882 g003
Figure 4. Summary of the interaction between γδ T cells and cancer cells.
Figure 4. Summary of the interaction between γδ T cells and cancer cells.
Cells 12 01882 g004
Figure 5. Illustration of eosinophils’ role in tumor microenvironment.
Figure 5. Illustration of eosinophils’ role in tumor microenvironment.
Cells 12 01882 g005
Figure 6. Summary of the influence of commensal microbiota in cancer immunity.
Figure 6. Summary of the influence of commensal microbiota in cancer immunity.
Cells 12 01882 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wen, M.; Li, Y.; Qin, X.; Qin, B.; Wang, Q. Insight into Cancer Immunity: MHCs, Immune Cells and Commensal Microbiota. Cells 2023, 12, 1882. https://doi.org/10.3390/cells12141882

AMA Style

Wen M, Li Y, Qin X, Qin B, Wang Q. Insight into Cancer Immunity: MHCs, Immune Cells and Commensal Microbiota. Cells. 2023; 12(14):1882. https://doi.org/10.3390/cells12141882

Chicago/Turabian Style

Wen, Minting, Yingjing Li, Xiaonan Qin, Bing Qin, and Qiong Wang. 2023. "Insight into Cancer Immunity: MHCs, Immune Cells and Commensal Microbiota" Cells 12, no. 14: 1882. https://doi.org/10.3390/cells12141882

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