Skip to main content
The Japanese Dental Science Review logoLink to The Japanese Dental Science Review
. 2019 Oct 7;55(1):113–120. doi: 10.1016/j.jdsr.2019.09.001

Immunomodulatory aspects in the progression and treatment of oral malignancy

Nobuo Kondoh a,, Masako Mizuno-Kamiya b, Naoki Umemura a, Eiji Takayama a, Harumi Kawaki a, Kenji Mitsudo c, Yasunori Muramatsu d, Shinichiro Sumitomo d
PMCID: PMC6806653  PMID: 31660091

Abstract

Inflammation substantially affects the risk of oral malignancy. Pro-inflammatory cytokine, interferon (IFN)-γ, confers anti-tumor activity using several different mechanisms. Conversely, higher expression of interleukin (IL)-17 is associated with worse prognosis. Monocyte chemotactic protein (MCP)-1 correlates positively with poor long-term survival of head and neck squamous cell carcinoma (HNSCC) patients. IL-1α affects cancer associated fibroblasts and macrophages, and promote several malignant phenotypes including immune suppression. Some anti-inflammatory cytokines, including IL-10 and transforming growth factor (TGF)-β, relate to pro-tumoral activities.

Among immune checkpoint modulators, programmed death (PD-)1 and PD-ligand (L)1 facilitate oral squamous cell carcinoma (OSCC) cell evasion from immune surveillance, and the expression status of these has a prognostic value.

OSCCs contain tumor associated macrophages (TAMs) as major stromal cells of their tumor microenvironment. Among the two distinctive states, M2 macrophages support tumor invasion, metastasis and immune suppression. Crosstalk between TAMs and OSCC or cancer-associated fibroblasts (CAF) plays an important role in the progression of OSCC.

Clinical trials with blocking antibodies against IL-1α or melanoma-associated antigens have been reported as therapeutic approaches against OSCCs. The most promising approach activating antitumor immunity is the blockade of PD-1/PD-L1 axis. Manipulating the polarization of pro-tumorigenic macrophages has been reported as a novel therapeutic approach.

Keywords: Immune suppression, Tumor microenvironment (TME), IL-1α, IFN-γ, Programmed death (PD)1, Programmed death ligand (PD-L)1, Tumor associated macrophage (TAM), Cancer-associated fibroblast (CAF)

1. Introduction

Immunomodulatory aspects of cancer tissues include several cancer cell-intrinsic and extrinsic mechanisms controlled in the tumor micro environment (TME), which promote progression and often confer resistance to their therapy [1]. The progression from premalignant lesions to OSCC is a complicated multi step process. Chronic inflammation can cause tissue damage and changes in the inflammatory cells and cytokines present in the TME. OSCCs could also be promoted by chronic inflammation occurred in the periodontitis. Chronic infection of resident microbiota P. gingivalis activates enzymatic cascades enhancing cellular invasion of OSCCs [2]. These changes promote the eventual development of tumors toward highly malignant phenotypes. The anti-inflammatory cytokines, such as IL-10 and TGF-β1, pro-inflammatory cytokines, including IFN-γ and some others, are specifically regulated under extrinsic and intrinsic mechanisms in tumor milieu [3]. Higher expression of IL-17 is associated with worse prognosis [4]. MCP-1 correlates positively with poor long-term survival of head and neck squamous cell carcinoma patients [5]. IL-1α from tumor cells specifically enhances the immune suppressive activity of mesenchymal cells [6].

On the other hand, the axis of immune check point inhibitors, represented by PD1/PD-L1, plays an important role in regulation of immune tolerance [7]. One of the main elements of the stromal cells, and contributing to the extracellular environment of solid tumors, is the TAMs, mostly polarizing to an M2 phenotype, that involve immune regulatory mechanisms leading to malignant metastatic distribution of OSCC [8].

Clinical trials with blocking antibodies against IL-1α, or vaccination against tumor-specific melanoma-associated antigens have been reported [9,10]. The most promising approach activating antitumor immunity is the blockade of the PD-1/PD-L1 axis. As novel therapeutic approaches manipulating the polarization of pro-tumorigenic macrophages using specific ligand to TLR3, bisphosphonate, and blockings of specific cytokines have been reported [[11], [12], [13], [14]].

In this review, we attempt to report immune suppressive mechanisms in the OSCC tissues, and also refer to effective or potential therapeutics against oral malignancy.

2. Function of inflammatory modulator

Epidemiological and molecular biological studies have revealed that inflammation substantially increases the risk of oral malignancy [2]. In fact, chronic inflammation can induce continuous tissue damage and can also induce specific inflammatory cytokines. Sun et al., have demonstrated that the expression of anti-inflammatory or pro-inflammatory cytokines (TGF-β1, IL-10, IL-4, or IFN-γ, MCP-1) are specifically regulated during development from premalignant lesions to OSCC tissues [3]. Helper T (Th) cells mainly participating in tumor immunology are functionally classified into Th1, Th2, and Th17 cells, according to secretory cytokines and immunological roles [15]. Th2 cytokines (IL-4, IL-5, and IL-10) are classified as anti-inflammatory ones, and are often related to pro-tumoral activities, whereas most Th1 cytokines, represented by IFN-γ, are classified as pro-inflammatory cytokines, and are associated with, in general, good prognosis [16]. Serum levels of IL-17A, TGF-β1, IL-4 and IL-10 were significantly higher in OSCC patients while IL-2 and IFN-γ were relatively low in OSCC patients when compared to controls [17]. Increased expression of IL-10 and TGF-β1, and decreased IFN-γ are associated with negative regulation of natural killer (NK) cells in OSCC patients [18]. Representative cytokines affecting inflammatory modification and tumor phenotypes are as follows.

2.1. Anti-inflammatory and pro-tumoral cytokines

IL-10 and TGF-β are representative anti-inflammatory and immunosuppressive cytokines that promote immune escape of neoplastic cells [[19], [20], [21], [22], [23]]. In fact, overexpression of IL-10 and TGF-β2 is associated with poorer prognosis of OSCCs [24]. IL-10 inhibits antigen depiction of antigen presenting cells (APCs), i.e. macrophages and dendritic cells [21], regulating differentiation of regulatory T cells [22], and conferring resistance to the action of cytotoxic T cell upon tumor cells [23]. Hypoxic stress induces several immune suppressive molecules including IL-10 and TGF-β that could induce the differentiation of tumor-associated macrophage into M2 type suppressing anti-tumor immunity [24]. Another anti-inflammatory cytokine, IL-4, is also considered to be pro-tumoral [3,16,17], however, this function may vary depending on the tumor’s diplotype [25].

2.2. IFN-γ

As a representative pro-inflammatory cytokine conferring anti-tumor activity, IFN-γ is mainly secreted by activated T cells and natural killer cells. It enhances macrophage activation, Th1/Th2 balance, cellular proliferation and apoptosis [26,27]. Hyper methylation of IFN-γ promoter has been denoted as an intrinsic mechanism for the down regulation of IFN-γ expression in macrophages infiltrating malignant OSCC tissues, rather than in benign and normal tissues. [28]. Interestingly, IFN-γ inhibits viability and migration of OSCC cells, and induces apoptosis, possibly by regulating ER stress and unfolded protein response (UPR) mechanisms [29]. The apoptosis induced by IFN-γ in head and neck SCC cell lines seems to be mediated by the activation of indoleamine 2,3 protein [30]. IFN-γ treatment of OSCC cells has also been revealed to downregulate heat shock protein 27, which is a proposed anti-apoptotic molecule [31]. Dentin sialophosphoprotein (DSPP) is expressed in the cytoplasm and perinuclear perimeter of OSCC cells, and the expression of this product is significantly elevated in poorly differentiated OSCC cells [32]. DSPP affects ER stress, the UPR and Ca homeostasis [29]. Treatment of OSCC cells with IFN-γ downregulates DSPP and matrix metalloproteinase (MMP-20), leading to disturbances in endoplasmic reticulum (ER) homeostasis, which may cause decreased cell viability, migration and increased apoptosis of OSCC cells [33]. Hypoxia-dependent pathways demonstrating HIF-1α play a key role in the development of OSCCs [34,35]. HIF-1α regulates CD4+ and CD8+ T cells survival, since in HIF-1-knockdown mice, activation of CD4+ and CD8+ T cells together with higher levels of IFN-γ production is observed [36].

2.3. IL-17

As a pro-inflammatory cytokine, IL-17 is primarily secreted by T-helper type 17 cells and neutrophils. Increased IL-17 expression has also been observed in OSCC [37]. IL-17 overexpression in tongue cancer is correlated with cancer progression [38]. Expression of IL-17 protein in the OSCC tissue is associated with worse prognosis, i.e. T classification, metastasis, clinical stages and recurrence. Interestingly the IL-17 protein is prominent at tumor invasion fronts (or budding sites) [4].

2.4. MCP-1/CCL2

Monocyte chemotactic protein 1 (MCP-1/ CCL2) is produced by fibroblasts, epithelial cells, monocytes, and various tumor cells. MCP-1 facilitates the recruitment of monocytes and macrophages into the local inflammatory tissues, where it regulates their functions [39] (see below section on tumor associated macrophage). MCP-1 correlates positively with upregulation of pro-survival signaling by Akt, ERK, and/or STAT, and results in poor long-term survival of HNSCC patients [5].

2.5. IL-1α

The IL-1 family consists of 11 molecules which control inflammation and immunity [40]. Several lines of evidence demonstrate pleiotropic roles of IL-1α in immune regulation in tumor milieu surrounding OSCCs. In OSCC tissues, IL-1α affects CAF or fibroblasts to produce C-C motif ligand (CCL) 7, C-X-C motif ligand (CXCL)1, IL-8 and CCL2, which promote motility and invasion of tumor cells as well as macrophages [[41], [42], [43], [44]]. In HNSCC patients, significant correlation between IL-1α expression and development of distant metastasis has been reported [45].

Immunosuppressive function of IL-1α via CAF has been reported in the tumor milieu of melanomas harboring BRAF (V600E) oncogene, by which the expression of IL-1α is specifically activated [46]. In the prostatic carcinoma tissue, IL-1α upregulates TGF-β secretion from mesenchymal stem cells, that is resulting in the suppression of immune cells, then induces the promotion of tumor cells [6].

As we have already reported, the immunosuppressive efficacy of the OSCC milieu in the early stage is developed without reliance on the induction of myeloid derived suppressor cells (MDSCs) [47]. In the early stage, as represented by mouse OSCC, Sq-1979 cells, IL-1α from OSCC cells can functionally enhance immune suppressive activity of mesenchymal stromal cells which are directly contacted with activated spleen cells [48]. In contrast, MDSCs are strongly induced in mice with metastasized, advanced OSCC cells [47]. In fact, IL-1 may specifically contribute to the development of early-stage OSCCs, since IL-1 receptor antagonist (IL1RN) is markedly downregulated in early OSCCs compared to premalignant lesions and advanced OSCCs [49]. The mechanism by which OSCCs differentially utilize immune modulation involving CAFs, MDSCs and several other factors is not fully understood. Among these aspects, IL-1α could be a promoter of motility, invasion and immune suppression in OSCC tissues. However, the function of IL-1α seems to be pleiotropic depending on the tumor cell types. On the other hand, tumor-derived IL-1β promotes tumor cell proliferation [50], or cancer cell invasion [51] in OSCCs; however, to date few reports note this molecule as an immunological modulator of oral malignancy.

Modification of inflammatory and tumoral phenotype by representative cytokines are summarized in Table 1.

Table 1.

Effects of cytokines upon inflammation and tumor phenotypes.

Cytokines Inflammaton Tumor References
IL-4 anti- pro- 3, 16, 17, 25
IL-5 anti- pro- 3, 16,
IL-10 anti- pro- 3, 16-20, 22-24
TGF-β1 anti- pro- 3, 17, 18, 21, 22, 24
IFN-γ pro- anti- 3, 17, 18, 26-28, 30, 31
IL-17 pro- pro- 17, 37, 38
MCP-1/CCL2 pro- (recruitment of monocyte) pro- 5, 39, 44
IL-1α pro- pro- 40-45, 48, 49

Promotive (pro-) and antithetic (anti-) effects are denoted.

3. Immune-checkpoint inhibitor

The regulation of T cell activity generally requires two signals. Firstly, mediation by T cell receptors which specifically recognize peptides presented by major histocompatibility complexes (MHCs) on antigen-presenting cells (APCs), and, secondly, CD28 as a receptor for T cell co-stimulatory molecules (also known as B7 family) ligands located on APCs [52]. The aberrant expression of B7 family molecules by tumor or stromal cells is known to contribute to the suppression of anti-tumor immunity. Among immune checkpoint modulators, PD-L1, FKBP51, B7-H4, B7-H6, ALHD1, B7-H3 and IDO1 are associated with poor OSCC patient prognosis, while CTLA-4, TLT-2, VISTA, PD-L2 and PD-1 seem not to have significant prognostic value [53].

3.1. PD-L1 and PD-L2 expression among the progressive malignancies

Immunological evasion of OSCC cells is strongly affected by the interaction between PD-L1/CD274 /B7-H1 and PD-1/CD279. PD-1 is the first identified as a type-I transmembrane receptor found in a murine T-cell hybridoma undergoing activation-induced cell death (AICD) [54]. The expression of PD-1 is prominent in the exhausted T cells. In several human tumors, a larger numbers of tumor infiltrating lymphocytes (TILs) express PD-1 and are well associated with disabled CD8+ T cell activity [55,56]. High levels of PD-1 expression correlate with increased tumor-infiltrating regulatory T (Treg) cells and reduced effector T cells; therefore, blocking PD-1 by specific antibody could effectively enhance anti-tumor immunity [57].

PD-L1 is a transmembrane glycoprotein belonging to the immunoglobulin (Ig) superfamily, and plays an integral role in the regulation of immune tolerance [58]. It is the main ligand of PD-1 and its expression is found not only on activated T cells, B cells, NK cells, macrophages, dendritic cells and mastocytes, but also on other non-immune cell types including several tumors [59,60]. Peripheral blood of lymph node metastasized (N+) OSCC patients shows significantly higher levels of PD-L1 mRNA than in N0 patients [61]. The mRNA of PD-L2, another ligand for PD1 [62] is also up-regulated in OSCC tissues compared to that of normal oral mucosa [63]. In OSCC tissues, a positive correlation was shown between PD-L1 expression and tumor size, lymph node metastasis and other malignant phenotypes [64,65]. However, among OSCC cells of different grades, PD-L1 expression is lower in high-grade invasive cell lines than in low-grade invasive OSCC cells; and inverse correlation is observed between PD-L1 expression and the degree of epithelial-mesenchymal transition (EMT) [65]. Similar observations have been reported in immunohistochemical analyses, revealing that levels of PD-L1 protein in OSCC cells positively correlate with favorable prognostic factors, i.e., elevated TIL levels, longer overall survival, smaller tumor size and lower lymph node metastasis [66]. These paradoxical observations can be accounted for by the fact that PD-L1 is not only expressed in the tumor cells, but also in stromal cells, i.e., tumor-associated macrophages, dendritic cells (DCs), and CAFs [67,68]. Though PD-L1 expression is lower in high-grade invasive OSCC cells, higher levels of PD-L1 expression in high-grade invasive OSCC tissues could be mainly attributable to stromal cells, for example macrophages and DCs [65].

3.2. Regulation of PD-1/PD-L1 in the tumor microenvironment

Expression of PD-L1 is controlled by extrinsic and intrinsic mechanisms. IL-2, IL-7 and IL-15 directly induce the expression of PD-1 on T cells as well as PD-L1 on APCs [69]. Transcription of PD-L1 can be strongly induced by IFN-γ in tumor cells [70]. TGF-β1 signaling is correlated with PD-L1 downregulation. Furthermore, TLR4-inhibitory peptide suppresses PD-L1 induction on macrophages and DCs co-cultured with mesenchymal-phenotype OSCC cells with a concomitant epithelial mesenchymal transition (EMT)-associated gene regulation [65]. These results suggest that the EMT-induced mechanism is critical for PD-L1 induction. The mediators for this cross talk between advanced OSCC cells and macrophages or DCs on PD-L1 expression should be elucidated.

EBV infection promotes latent membrane protein 1 (LMP1) expression on the membrane of tumor cells, then LMP1 stimulates PD-L1 expression through STAT3, AP-1 and NF-KB signaling pathways [71]. This may promote immunotolerance in the infected cells. MicroRNAs (miRNAs) are single-stranded, noncoding short RNAs that inhibit the expression of genes. Some micro RNAs are correlated with progression of OSCC [72]. High miR-197 expression is tightly correlated with lower overall survival and reduced PD-L1 transcription and TIL in OSCC tissues, demonstrating that PD-L1 expression is regulated by miR-197 in OSCC cells [68].

The phosphatidylinositol-3-OH kinase (PI3K) (also known AKT-mTOR) signaling pathway is essential for both cell growth and survival, and is negatively regulated by a tumor suppressor gene product, phosphatase and tensin homolog (PTEN) [73]. Oncogenic activation of the AKT–mTOR pathway promotes immunological evasion by stimulating expression of PD-L1 in non-small cell lung cancer [74]. PD-L1 is post transcriptionally enhanced after loss of PTEN and activation of the PI3K pathway in glioma [75]. The expression level of PTEN was significantly lower in OSCC specimens when compared with adjacent normal tissues [76,77]. Interestingly, down regulations of PTEN in OSCCs are mediated by non-coding micro RNAs, including miR-221/222, miR-21 and miR-1297 [[78], [79], [80], [81]]. Thus, the PTEN/PD-L1 axis may be a potential target for immune-checkpoint therapy against OSCC.

Hypoxic stress induces several immune suppressive molecules. The binding of HIF-1α to the hypoxia-response element of the PD-L1 promoter may lead to resistance to CTL-mediated cell lysis [82]. TAMs in hypoxic regions could upregulate PD-L1 expression via HIF-1α and induce T-cell suppression [83,84]. Interestingly, in progressive OSCCs, both co-localized HIF-1a and PD-L1 are associated with worse prognosis [85].

4. Tumor associated macrophages

4.1. Specific macrophage polarization

It is known that the majority of malignant tumors contain macrophages as major stromal cells of their tumor microenvironment. Unlike macrophages in normal tissue, TAMs are modified in the tumor milieu, with some losing the ability to phagocytize or present tumor antigens to T-cells [86]. TAMs harbor two distinct phenotypes: one is the conventionally activated (M1) state and the other is the differently activated (M2) state, with these two states representing the extreme polarized phenotypes [87,88]. The M1 polarization state depends on microbial stimulus, represents T helper type 1 (TH1) cytokine profiles and possesses antitumor activity [89]. Interestingly, M1 macrophages were observed to select CD47 negative OSCC cells for phagocytosis, although M2 did not, indicating that the expression of CD47 on OSCC cells could be a marker for evasion from M1 phagocytosis [90]. On the other hand, immune suppressive M2 polarization depends on T helper type 2 (TH2) cytokine profiles [88]. M2 macrophages harbor heterogeneous phenotypes that support tumor progression including invasion, metastasis and immune suppression [91]. Larger numbers of M2 macrophages were closely related to shorter survival times [90]. CD68 is considered to be a pan-macrophage marker, CD163 is an M2 marker and CD11c is an M1 marker [92]. Higher concentrations of CD163 and CD68 in OSCC tissues, were markedly correlated with worse survival rates [92]. Tumor immunology is strongly associated with lymph node metastasis. Immune tolerance in local lymph nodes is a prerequisite for lymph node metastasis. In higher grade OSCC patients, a shift of macrophage polarization from M1 to M2 occurs even in the sinuses of unmetastasized lymph nodes [93,94]. This immunological tolerance is associated with high galectin 3 expression in tumor-free lymph nodes [95]. In higher grade OSCCs, TAMs are further subdivided; CD163+CD204+ TAMs strongly produce IL-10 and PD-L1 and also reduce CD3+ T cells in comparison with CD163+CD204- and CD163-CD204+ TAMs. CD163+CD204+ TAMs negatively correlate with the population of activated (CD25+) T cells and 5-year progression-free survival [96].

4.2. Crosstalk among cancer and stromal cells

Crosstalk between TAMs and tumor cells is thought to play an important role in OSCC. Increasing expression of kinesin family member 4A (Kif4A) and high infiltration of M2 macrophages in both OSCC and macrophages, causing the overproduction of chemokine C—C motif ligand 2 (CCL2/ MCP-1) and CCR, have been correlated with greater tumor size and poor prognosis [97]. Notably, CCL2 promotes chemotaxis of M0 or M2 macrophages and induces M2 phenotype polarization [98]. Therefore, the Kif4A-CCL2 /CCR2-macrophage axis could be accounted as a novel therapeutic target. MCP-1 mediates osteoclastogenesis of mononuclear cells, and promotes osteoclast development in cancers [99]. Furthermore, a deletion mutant of MCP-1 inhibits osteoclast differentiation of stromal monocytes and reduces bone invasion of OSCC cells [14]. Overexpression of leukocyte-associated immunoglobulin-like receptor (LAIR)-1 in OSCC cells is associated with advanced pathological grade and correlates with immune suppressive features of the tumor tissue; in fact, the expression is tightly associated with immune suppressive markers of TAM and MDSC, i.e., CD11b, CD163, CD33, CD68, two immune checkpoints (B7-H3 and VISTA) and indoleamine 2,3-dioxygenase (IDO) [100]. OSCC suppresses antitumor immunity by the induction of PD-L1 on M2 type TAMs; the IL-10 concentration in the tumor microenvironment directly correlates with the PD-L1 level on the TAMs [101]. Interestingly, TLR4-inhibitory peptide successfully suppressed PD-L1 upregulation on TAMs co-cultured with OSCC cells, suggesting that some EMT-inducing tumor antigen(s) is critical for PD-L1 induction via TLR4 on the cells [65]. 

Crosstalk with CAFs is also essential to develop TAMs. CAFs are classified into 3 grades depending on the expression of alpha smooth muscle actin, and the CAFs in the higher grade promote the development of CD163 positive macrophages, and promote poor prognosis in OSCC tissues [102]. CAFs educate macrophage progenitor cells via TGF-β1, IL-10 and ARG1, then induce the protumoral TAMs in the tumor immunosuppressive milieu [103].

5. Therapeutic approaches

In the development and progression of OSCCs, immune system plays very important roles. Here we report potential cancer immunotherapies targeting the molecules in the tumor microenvironment including tumor-specific molecules, immune check point inhibitor or TAMs.

5.1. Targeting tumor-specific molecules

Clinical trials with an IL-1α-blocking monoclonal antibody has shown promising outcomes in patients with advanced cancers [9,104]. MABp1 (XBiotech Inc.) is a naturally occurring human IL-1α neutralizing antibody. Monotherapy with MABp1 improved survival rate and other clinical improvements in patients with advanced non-small cell lung cancer, ovarian cancer and other refractory cancers [9,105].

Melanoma-associated antigen (MAGE) genes are expressed in a variety of neoplastic lesions including breast, lung and oral cancers, and are considered to be potential targets of vaccination for treatment [[106], [107], [108]]. This gene family is classified into acidic subgroups, MAGE-A, -B, -C, and a basic MAGE-D [109]. The expression of MAGED4B is elevated in more than 50% of OSCC tissues. Interestingly, the overexpression of this molecule promotes cell migration, cell growth, and conferred resistance to apoptosis [110]. Synthetic peptides of MAGED4B have binding affinity against the MHC-Class I molecules and enhacned IFN-γ and Granzyme-B production in blood cells from OSCC patients, demonstrating that they are immunogenic; furthermore the peptide-pulsed dendritic cells enhance T-cell cytotoxicity against MAGED4B-overexpressing OSCC cell lines, demonstrating that they could be promising candidates of vaccination for OSCC treatment [10]. In OSCCs, the expression of MAGE A subfamilies A1-A12 is significantly associated with their malignancy and could be potential targets for cancer immunotherapy [111].

5.2. Targeting immune checkpoint inhibitors

Recently, the most promising approach in activating therapeutic antitumor immunity is the blockade of immune checkpoints. In clinical trials, therapeutic antibodies against PD-1 [112], PD-L1 [113], and in combination against both cytotoxic T-lymphocyte-associated protein (CTLA)-4 and PD-L1 [114] have been shown to extend the survival of many cancer patients. A clinical trial in phase I study of OSCC patients using novel anti-PD-1 antibody is also reported [115]. Expression of PD-L1 by tumor cells and immune infiltrates is markedly associated with the expression of PD-1 on lymphocytes, as well as responses to anti-PD-1 therapy [116], or anti-PD-L1 therapy [117]. The PD-1 blockade is accompanied by induction of IFN-γ, STAT-1 activation and the production of the T cell effector granzyme B. This activity prevents the development of carcinogen-induced oral premalignant lesions [118].

As a novel approach, exosomal delivery of miRNA targeting immune checkpoint inhibitor has been reported. γδ T cell-derived extracellular vesicles (γδTDEs) delivering miR-138 efficiently targets PD-1 and CTLA-4 in CD8+ T cells, then activates them to achieve synergistic therapeutic effects on OSCC [119].

5.3. Targeting TAMs

Manipulating polarization of pro-tumorigenic M2 to the anti-tumor M1 macrophage phenotype provides a new therapeutic approach. Stimulation of TLR3/Toll-IL-1 receptor domain-containing adaptor molecule 1 using Poly (I:C) immediately enhances the secretion of several proinflammatory cytokines, including IL-1β, and accelerates M1 macrophage polarization [11]. The TLR3 agonist Poly(I:C) is effective in triggering the cytotoxic activity of tumor conditioned macrophage against cancer cells [120].

Another approach targeting macrophage polarization could be challenged using bisphosphonates. Bisphosphonates have cytotoxic potential on myeloid cells and are useful for the treatment of osteoporosis and prevention of bone metastases. Osteoclast stimulatory transmembrane protein could induce a phenotypic switch in macrophage polarization [121]. Zoledronic acid promotes TLR-4-mediated M1 macrophage polarization [122]. Several strategies aiming at re-education and/or depletion of M2-like protumoral TAM, including drug delivery of bisphosphonates, are reported [12].

Macrophage colony-stimulating factor (CSF-1) is a potential target in preventing macrophage recruitment. In animal models, CSF-1 receptor inhibition strongly reduces tumor-associated macrophages and increase the CD8+/CD4+ T cell ratio [13]. The dominant-negative deletion-mutant of MCP-1 inhibits osteoclast differentiation of monocyte and reduces the bone invasion by OSCC cells in vivo [14]. The blockade of MCP-1 could also prevent M2 phenotype polarization of macrophages.

5.4. For improvement of surgical treatment

Surgical treatment is still the most effective method of removing primary and metastasized OSCCs. Anesthetic choice may affect immune regulation and prognostic implications. Lydokine increases NK cell activity. Anesthetics using propofol decrease neuroendocrine response, and may cause less immunosuppression when compared to volatile anesthetics and opioids [123]. In general, anesthetics reduce IL-1, while enhancing IL-10 levels [123]. They may modulate surgical stress by acting on the central nervous system, affecting catecholamines and glucocorticoids [124].

6. Conclusion

There are several immune modulators, including cytokines, immune-check point inhibitors, and cellular components in the TME of OSCC. These molecules and cellular components are regulated by extrinsic and intrinsic mechanisms among the TME. There are several therapeutic approaches targeting these activities using specific antibodies, miRNAs and T cell-derived extracellular vesicles. As a novel therapeutic approach, alternative differentiation of TAM could be induced using bisphosphonates. However, when considering more fine tuning of therapeutic approaches, pro-tumoral immune components are not fully understood. Linking of anesthesiology and immunology may provide new therapeutic advantages. However, the mechanisms by which anesthetic drugs affect the immune system remains unclear.

Further elucidation of the regulatory mechanisms consist of tumor-host interactions could identify important therapeutic targets in OSCC development.

Conflict of interest

None.

Acknowledgements

This work was financially supported in part by the Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (KAKENHI) Grant Number (JP17K11891).

References

  • 1.Sharma P., Hu-Lieskovan S., Wargo J.A., Ribas A. Primary, adaptive, and acquired resistance to Cancer immunotherapy. Cell. 2017;168(4):707–723. doi: 10.1016/j.cell.2017.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Inaba H., Sugita H., Kuboniwa M., Iwai S., Hamada M., Noda T. Porphyromonas gingivalis promotes invasion of oral squamous cell carcinoma through induction of proMMP9 and its activation. Cell Microbiol. 2014;16(1):131–145. doi: 10.1111/cmi.12211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Sun Y., Liu N., Guan X., Wu H., Sun Z., Zeng H. Immunosuppression induced by chronic inflammation and the progression to oral squamous cell carcinoma. Mediators Inflamm. 2016;2016 doi: 10.1155/2016/5715719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zhang S., Wang X., Gupta A., Fang X., Wang L., Zhang C. Expression of IL-17 with tumor budding as a prognostic marker in oral squamous cell carcinoma. Am J Transl Res. 2019;11(3):1876–1883. [PMC free article] [PubMed] [Google Scholar]
  • 5.Ji W.T., Chen H.R., Lin C.H., Lee J.W., Lee C.C. Monocyte chemotactic protein 1 (MCP-1) modulates pro-survival signaling to promote progression of head and neck squamous cell carcinoma. PLoS One. 2014;9(2):e88952. doi: 10.1371/journal.pone.0088952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cheng J., Li L., Liu Y., Wang Z., Zhu X., Bai X. Interleukin-1α induces immunosuppression by mesenchymal stem cells promoting the growth of prostate cancer cells. Mol Med Rep. 2012;6(5):955–960. doi: 10.3892/mmr.2012.1019. [DOI] [PubMed] [Google Scholar]
  • 7.Lenouvel D., González-Moles M.Á, Talbaoui A., Ramos-García P., González-Ruiz L., Ruiz-Ávila I. An update of knowledge on PD-L1 in head and neck cancers: physiologic, prognostic and therapeutic perspectives. Oral Dis. 2019 doi: 10.1111/odi.13088. March 13. [DOI] [PubMed] [Google Scholar]
  • 8.Petruzzi M.N., Cherubini K., Salum F.G., de Figueiredo M.A. Role of tumour-associated macrophages in oral squamous cells carcinoma progression: an update on current knowledge. Diagn Pathol. 2017;12(1):32. doi: 10.1186/s13000-017-0623-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hong D.S., Hui D., Bruera E., Janku F., Naing A., Falchook G.S. MABp1, a first-in-class true human antibody targeting interleukin-1α in refractory cancers: an open-label, phase 1 dose-escalation and expansion study. Lancet Oncol. 2014;15(6):656–666. doi: 10.1016/S1470-2045(14)70155-X. [DOI] [PubMed] [Google Scholar]
  • 10.Lim K.P., Chun N.A., Gan C.P., Teo S.H., Rahman Z.A., Abraham M.T. Identification of immunogenic MAGED4B peptides for vaccine development in oral cancer immunotherapy. Hum Vaccin Immunother. 2014;10(11):3214–3223. doi: 10.4161/hv.29226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Shime H., Matsumoto M., Oshiumi H., Tanaka S., Nakane A., Iwakura Y. Toll-like receptor 3 signaling converts tumor-supporting myeloid cells to tumoricidal effectors. Proc Natl Acad Sci U S A. 2012;109(6):2066–2071. doi: 10.1073/pnas.1113099109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ponzoni M., Pastorino F., Di Paolo D., Perri P., Brignole C. Targeting macrophages as a potential therapeutic intervention: impact on inflammatory diseases and cancer. Int J Mol Sci. 2018;19(7) doi: 10.3390/ijms19071953. pii: E1953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ries C.H., Cannarile M.A., Hoves S., Benz J., Wartha K., Runza V. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell. 2014;25(6):846–859. doi: 10.1016/j.ccr.2014.05.016. [DOI] [PubMed] [Google Scholar]
  • 14.Luo S., Zhou C., Zhang J., Chen M., Li H., Zheng S. Mutant monocyte chemoattractant protein-1 protein (7ND) inhibits osteoclast differentiation and reduces oral squamous carcinoma cell bone invasion. Oncol Lett. 2018;15(5):7760–7768. doi: 10.3892/ol.2018.8308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhou L., Chong M.M., Littman D.R. Plasticity of CD4+ T cell lineage differentiation. Immunity. 2009;30(5):646–655. doi: 10.1016/j.immuni.2009.05.001. [DOI] [PubMed] [Google Scholar]
  • 16.Mantovani A., Allavena P., Sica A., Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–444. doi: 10.1038/nature07205. [DOI] [PubMed] [Google Scholar]
  • 17.Gaur P., Singh A.K., Shukla N.K., Das S.N. Inter-relation of Th1, Th2, Th17 and Treg cytokines in oral cancer patients and their clinical significance. Hum Immunol. 2014;75(4):330–337. doi: 10.1016/j.humimm.2014.01.011. [DOI] [PubMed] [Google Scholar]
  • 18.Dutta A., Banerjee A., Saikia N., Phookan J., Baruah M.N., Baruah S. Negative regulation of natural killer cell in tumor tissue and peripheral blood of oral squamous cell carcinoma. Cytokine. 2015;76(2):123–130. doi: 10.1016/j.cyto.2015.09.006. [DOI] [PubMed] [Google Scholar]
  • 19.Matsuda M., Salazar F., Petersson M., Masucci G., Hansson J., Pisa P. Interleukin 10 pretreatment protects target cells from tumor- and allo-specific cytotoxic T cells and downregulates HLA class I expression. J Exp Med. 1994;180(6):2371–2376. doi: 10.1084/jem.180.6.2371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kryczek I., Wei S., Zou L., Zhu G., Mottram P., Xu H. Cutting edge: induction of B7-H4 on APCs through IL-10: novel suppressive mode for regulatory T cells. J Immunol. 2006;177(1):40–44. doi: 10.4049/jimmunol.177.1.40. [DOI] [PubMed] [Google Scholar]
  • 21.Thomas D.A., Massagué J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell. 2005;8(5):369–380. doi: 10.1016/j.ccr.2005.10.012. [DOI] [PubMed] [Google Scholar]
  • 22.Arantes D.A., Costa N.L., Mendonça E.F., Silva T.A., Batista A.C. Overexpression of immunosuppressive cytokines is associated with poorer clinical stage of oral squamous cell carcinoma. Arch Oral Biol. 2016;61:28–35. doi: 10.1016/j.archoralbio.2015.10.013. [DOI] [PubMed] [Google Scholar]
  • 23.Kurte M., López M., Aguirre A., Escobar A., Aguillón J.C., Charo J. A synthetic peptide homologous to functional domain of human IL-10 down-regulates expression of MHC class I and Transporter associated with Antigen Processing 1/2 in human melanoma cells. J Immunol. 2004;173(3):1731–1737. doi: 10.4049/jimmunol.173.3.1731. [DOI] [PubMed] [Google Scholar]
  • 24.Hao N.B., Lü M.H., Fan Y.H., Cao Y.L., Zhang Z.R., Yang S.M. Macrophages in tumor microenvironments and the progression of tumors. Clin Dev Immunol. 2012;2012 doi: 10.1155/2012/948098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yang C.M., Chen H.C., Hou Y.Y., Lee M.C., Liou H.H., Huang S.J. A high IL-4 production diplotype is associated with an increased risk but better prognosis of oral and pharyngeal carcinomas. Arch Oral Biol. 2014;59(1):35–46. doi: 10.1016/j.archoralbio.2013.09.010. [DOI] [PubMed] [Google Scholar]
  • 26.Gattoni A., Parlato A., Vangieri B., Bresciani M., Derna R. Interferon-gamma: biologic functions and HCV therapy (type I/II) (2 of 2 parts) Clin Ter. 2006;157(5):457–468. [PubMed] [Google Scholar]
  • 27.Chawla-Sarkar M., Lindner D.J., Liu Y.F., Williams B.R., Sen G.C., Silverman R.H. Apoptosis and interferons: role of interferon-stimulated genes as mediators of apoptosis. Apoptosis. 2003;8:237–249. doi: 10.1023/a:1023668705040. [DOI] [PubMed] [Google Scholar]
  • 28.Tian S., Jiang C., Liu X., Xu S., Zhang Z., Chen H. Hypermethylation of IFN-γ in oral cancer tissues. Clin Oral Investig. 2017;21(8):2535–2542. doi: 10.1007/s00784-017-2052-z. [DOI] [PubMed] [Google Scholar]
  • 29.Gkouveris I., Nikitakis N.G., Aseervatham J., Ogbureke K.U.E. The tumorigenic role of DSPP and its potential regulation of the unfolded protein response and ER stress in oral cancer cells. Int J Oncol. 2018;53:1743–1751. doi: 10.3892/ijo.2018.4484. [DOI] [PubMed] [Google Scholar]
  • 30.El Jamal S.M., Taylor E.B., Abd Elmageed Z.Y., Alamodi A.A., Selimovic D., Alkhateeb A. Interferon gamma-induced apoptosis of head and neck squamous cell carcinoma is connected to indoleamine-2,3-dioxygenase via mitochondrial and ER stress‑associated pathways. Cell Div. 2016;11:11. doi: 10.1186/s13008-016-0023-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yonekura N., Yokota S., Yonekura K., Dehari H., Arata S., Kohama Interferon-gamma downregulates Hsp27 expression and suppresses the negative regulation of cell death in oral squamous cell carcinoma lines. Cell Death Differ. 2003;10(March (3)):313–322. doi: 10.1038/sj.cdd.4401169. [DOI] [PubMed] [Google Scholar]
  • 32.Ogbureke K.U., Nikitakis N.G., Warburton G., Ord R.A., Sauk J.J., Waller J.L. Up-regulation of SIBLING proteins and correlation with cognate MMP expression in oral cancer. Oral Oncol. 2007;43:920–932. doi: 10.1016/j.oraloncology.2006.11.011. [DOI] [PubMed] [Google Scholar]
  • 33.Gkouveris I., Nikitakis N.G., Aseervatham J., Ogbureke K.U.E. Interferon γ suppresses dentin sialophosphoprotein in oral squamous cell carcinoma cells resulting in antitumor effects, via modulation of the endoplasmic reticulum response. Int J Oncol. 2018;53(6):2423–2432. doi: 10.3892/ijo.2018.4590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Eckert A.W., Kappler M., Schubert J., Taubert H. Correlation of expression of hypoxia-related proteins with prognosis in oral squamous cell carcinoma patients. Oral Maxillofac Surg. 2012;16(2):189–196. doi: 10.1007/s10006-012-0335-8. [DOI] [PubMed] [Google Scholar]
  • 35.Gong L., Zhang W., Zhou J., Lu J., Xiong H., Shi X. Prognostic value of HIFs expression in head and neck cancer: a systematic review. PLoS One. 2013;8(9) doi: 10.1371/journal.pone.0075094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lukashev D., Klebanov B., Kojima H., Grinberg A., Ohta A., Berenfeld L. Cutting edge: hypoxia-inducible factor 1alpha and its activation-inducible short isoform I.1 negatively regulate functions of CD4+ and CD8+ T lymphocytes. J Immunol. 2006;177(8):4962–4965. doi: 10.4049/jimmunol.177.8.4962. [DOI] [PubMed] [Google Scholar]
  • 37.Avadhani A.V., Parachuru V.P., Milne T., Seymour G.J., Rich A.M. Multiple cells express interleukin 17 in oral squamous cell carcinoma. J Oral Pathol Med. 2017;46(1):39–45. doi: 10.1111/jop.12465. [DOI] [PubMed] [Google Scholar]
  • 38.Wei T., Cong X., Wang X.T., Xu X.J., Min S.N., Ye P. Interleukin-17A promotes tongue squamous cell carcinoma metastasis through activating miR-23b/versican pathway. Oncotarget. 2017;8(4):6663–6680. doi: 10.18632/oncotarget.14255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Deshmane S.L., Kremlev S., Amini S., Sawaya B.E. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. 2009;29(6):313–326. doi: 10.1089/jir.2008.0027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dinarello C.A. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27:519–550. doi: 10.1146/annurev.immunol.021908.132612. [DOI] [PubMed] [Google Scholar]
  • 41.Bae J.Y., Kim E.K., Yang D.H., Zhang X., Park Y.J., Lee D.Y. Reciprocal interaction between carcinoma-associated fibroblasts and squamous carcinoma cells through interleukin-1α induces cancer progression. Neoplasia. 2014;16(11):928–938. doi: 10.1016/j.neo.2014.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jung D.W., Che Z.M., Kim J., Kim K., Kim K.Y., Williams D. Tumor-stromal crosstalk in invasion of oral squamous cell carcinoma: a pivotal role of CCL7. Int J Cancer. 2010;127(2):332–344. doi: 10.1002/ijc.25060. [DOI] [PubMed] [Google Scholar]
  • 43.Grimm S., Jennek S., Singh R., Enkelmann A., Junker K., Rippaus N. Malignancy of bladder cancer cells is enhanced by tumor-associated fibroblasts through a multifaceted cytokine-chemokine loop. Exp Cell Res. 2015;335(1):1–11. doi: 10.1016/j.yexcr.2015.04.001. [DOI] [PubMed] [Google Scholar]
  • 44.Paish H.L., Kalson N.S., Smith G.R., Del Carpio Pons A., Baldock T.E., Smith N. Fibroblasts promote inflammation and pain via IL-1α induction of the monocyte chemoattractant chemokine (C-C motif) ligand 2. Am J Pathol. 2018;188(3):696–714. doi: 10.1016/j.ajpath.2017.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.León X., Bothe C., García J., Parreño M., Alcolea S., Quer M. Expression of IL-1α correlates with distant metastasis in patients with head and neck squamous cell carcinoma. Oncotarget. 2015;6(35):37398–37409. doi: 10.18632/oncotarget.6054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Khalili J.S., Liu S., Rodríguez-Cruz T.G., Whittington M., Wardell S., Liu C. Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clin Cancer Res. 2012;18(19):5329–5340. doi: 10.1158/1078-0432.CCR-12-1632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kondoh N., Mizuno-Kamiya M., Takayama E., Kawati H., Umemura N., Yamazaki Y. Perspectives of immune suppression in the tumor microenvironment promoting oral malignancy. Open Dent J. 2018;12:455–465. doi: 10.2174/1874210601812010455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Morimoto-Ito H., Mizuno-Kamiya M., Umemura N., Inagaki Y., Takayama E., Kawaki H. Immunosuppressive effect of mesenchymal stromal cells is enhanced by IL-1α from oral squamous cell carcinoma cells. Open Dent J. 2019;13(1):221–227. [Google Scholar]
  • 49.Shiiba M., Saito K., Yamagami H., Nakashima D., Higo M., Kasamatsu A. Interleukin-1 receptor antagonist (IL1RN) is associated with suppression of early carcinogenic events in human oral malignancies. Int J Oncol. 2015;46(5):1978–1984. doi: 10.3892/ijo.2015.2917. [DOI] [PubMed] [Google Scholar]
  • 50.Lee M.K., Park J.H., Gi S.H., Hwang Y.S. IL-1β induces fascin expression and increases Cancer invasion. Anticancer Res. 2018;38(11):6127–6132. doi: 10.21873/anticanres.12964. [DOI] [PubMed] [Google Scholar]
  • 51.Wu J., Hong Y., Wu T., Wang J., Chen X., Wang Z. Stromal-epithelial lactate shuttle induced by tumor-derived interleukin-1β promotes cell proliferation in oral squamous cell carcinoma. Int J Mol Med. 2018;41(2):687–696. doi: 10.3892/ijmm.2017.3267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ceeraz S., Nowak E.C., Noelle R.J. B7 family checkpoint regulators in immune regulation and disease. Trends Immunol. 2013;34(11):556–563. doi: 10.1016/j.it.2013.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sieviläinen M., Almahmoudi R., Al-Samadi A., Salo T., Pirinen M., Almangush A. The prognostic value of immune checkpoints in oral squamous cell carcinoma. Oral Dis. 2018 doi: 10.1111/odi.12991. October 13 [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 54.Ishida Y., Agata Y., Shibahara K., Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11(11):3887–3895. doi: 10.1002/j.1460-2075.1992.tb05481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ahmadzadeh M., Johnson L.A., Heemskerk B., Wunderlich J.R., Dudley M.E., White D.E. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114(8):1537–1544. doi: 10.1182/blood-2008-12-195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Matsuzaki J., Gnjatic S., Mhawech-Fauceglia P., Beck A., Miller A., Tsuji T. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci U S A. 2010;107(17):7875–7880. doi: 10.1073/pnas.1003345107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dyck L., Wilk M.M., Raverdeau M., Misiak A., Boon L., Mills K.H. Anti-PD-1 inhibits Foxp3(+) Treg cell conversion and unleashes intratumoural effector T cells thereby enhancing the efficacy of a cancer vaccine in a mouse model. Cancer Immunol Immunother. 2016;65(12):1491–1498. doi: 10.1007/s00262-016-1906-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4(5):336–347. doi: 10.1038/nri1349. [DOI] [PubMed] [Google Scholar]
  • 59.Sharpe A.H., Wherry E.J., Ahmed R., Freeman G.J. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8(3):239–245. doi: 10.1038/ni1443. [DOI] [PubMed] [Google Scholar]
  • 60.Badoual C., Hans S., Merillon N., Van Ryswick C., Ravel P., Benhamouda N. PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res. 2013;73(1):128–138. doi: 10.1158/0008-5472.CAN-12-2606. [DOI] [PubMed] [Google Scholar]
  • 61.Weber M., Wehrhan F., Baran C., Agaimy A., Büttner-Herold M., Preidl R. PD-L1 expression in tumor tissue and peripheral blood of patients with oral squamous cell carcinoma. Oncotarget. 2017;8(68):112584–112597. doi: 10.18632/oncotarget.22576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bardhan K., Anagnostou T., Boussiotis V.A. The PD1:PD-L1/2 pathway from discovery to clinical implementation. Front Immunol. 2016;7:550. doi: 10.3389/fimmu.2016.00550. December 12, eCollection 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Weber M., Wehrhan F., Baran C., Agaimy A., Büttner-Herold M., Kesting M. Prognostic significance of PD-L2 expression in patients with oral squamous cell carcinoma—a comparison to the PD-L1 expression profile. Cancer Med. 2019;8(3):1124–1134. doi: 10.1002/cam4.1929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Oliveira-Costa J.P., de Carvalho A.F., da Silveira da G.G., Amaya P., Wu Y., Park K.J. Gene expression patterns through oral squamous cell carcinoma development: PD-L1 expression in primary tumor and circulating tumor cells. Oncotarget. 2015;6(25):20902–20920. doi: 10.18632/oncotarget.3939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hirai M., Kitahara H., Kobayashi Y., Kato K., Bou-Gharios G., Nakamura H. Regulation of PD-L1 expression in a high-grade invasive human oral squamous cell carcinoma microenvironment. Int J Oncol. 2017;50(1):41–48. doi: 10.3892/ijo.2016.3785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ahn H., Yang J.M., Kim H., Chung J.H., Ahn S.H., Jeong W.J. Clinicopathologic implications of the miR-197/PD-L1 axis in oral squamous cell carcinoma. Oncotarget. 2017;8(39):66178–66194. doi: 10.18632/oncotarget.19842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Okazaki T., Chikuma S., Iwai Y., Fagarasan S., Honjo T. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat Immunol. 2013;14(12):1212–1218. doi: 10.1038/ni.2762. [DOI] [PubMed] [Google Scholar]
  • 68.Nazareth M.R., Broderick L., Simpson-Abelson M.R., Kelleher R.J., Jr, Yokota S.J., Bankert R.B. Characterization of human lung tumor-associated fibroblasts and their ability to modulate the activation of tumor-associated T cells. J Immunol. 2007;178(9):5552–5562. doi: 10.4049/jimmunol.178.9.5552. [DOI] [PubMed] [Google Scholar]
  • 69.Kinter A.L., Godbout E.J., McNally J.P., Sereti I., Roby G.A., O’Shea M.A. The common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J Immunol. 2008;181(10):6738–6746. doi: 10.4049/jimmunol.181.10.6738. [DOI] [PubMed] [Google Scholar]
  • 70.Dong H., Strome S.E., Salomao D.R., Tamura H., Hirano F., Flies D.B. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi: 10.1038/nm730. [DOI] [PubMed] [Google Scholar]
  • 71.Fang W., Zhang J., Hong S., Zhan J., Chen N., Qin T. EBV-driven LMP1 and IFN-γ up-regulate PD-L1 in nasopharyngeal carcinoma: implications for oncotargeted therapy. Oncotarget. 2014;5(23):12189–12202. doi: 10.18632/oncotarget.2608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Soga D., Yoshiba S., Shiogama S., Miyazaki H., Kondo S., Shintani S. microRNA expression profiles in oral squamous cell carcinoma. Oncol Rep. 2013;30(2):579–583. doi: 10.3892/or.2013.2488. [DOI] [PubMed] [Google Scholar]
  • 73.Downes C.P., Ross S., Maccario H., Perera N., Davidson L., Leslie N.R. Stimulation of PI 3-kinase signaling via inhibition of the tumor suppressor phosphatase, PTEN. Adv Enzyme Regul. 2007;47:184–194. doi: 10.1016/j.advenzreg.2006.12.018. [DOI] [PubMed] [Google Scholar]
  • 74.Lastwika K.J., Wilson W., 3rd, Li Q.K., Norris J., Xu H., Ghazarian S.R. Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res. 2016;76(2):227–238. doi: 10.1158/0008-5472.CAN-14-3362. [DOI] [PubMed] [Google Scholar]
  • 75.Parsa A.T., Waldron J.S., Panner A., Crane C.A., Parney I.F., Barry J.J. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007;13(1):84–88. doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
  • 76.Baghaei F., Abdollahi A., Mohammadpour H., Jahanbin M., Naseri Taheri F. PTEN and miR-26b: promising prognostic biomarkers in initiation and progression of oral squamous cell carcinoma. J Oral Pathol Med. 2019;48(1):31–35. doi: 10.1111/jop.12794. [DOI] [PubMed] [Google Scholar]
  • 77.Jasphin S.S., Desai D., Pandit S., Gonsalves N.M., Nayak P.B., Iype A. Immunohistochemical expression of phosphatase and tensin homolog in histologic gradings of oral squamous cell carcinoma. Contemp Clin Dent. 2016;7(4):524–528. doi: 10.4103/0976-237X.194111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zhou L., Jiang F., Chen X., Liu Z., Ouyang Y., Zhao W. Downregulation of miR-221/222 by a microRNA sponge promotes apoptosis in oral squamous cell carcinoma cells through upregulation of PTEN. Oncol Lett. 2016;12(6):4419–4426. doi: 10.3892/ol.2016.5250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gao L., Ren W., Zhang L., Li S., Kong X., Zhang H. PTENp1, a natural sponge of miR-21, mediates PTEN expression to inhibit the proliferation of oral squamous cell carcinoma. Mol Carcinog. 2017;56(4):1322–1334. doi: 10.1002/mc.22594. [DOI] [PubMed] [Google Scholar]
  • 80.Patel S., Rawal R. Role of miRNA dynamics and cytokine profile in governing CD44v6/Nanog/PTEN axis in oral cancer: modulating the master regulators. Tumour Biol. 2016;37(11):14565–14575. doi: 10.1007/s13277-016-5289-2. [DOI] [PubMed] [Google Scholar]
  • 81.Liang L., Feng L., Wei B. microRNA-1297 involves in the progression of oral squamous cell carcinoma through PTEN. Saudi J Biol Sci. 2018;25(5):923–927. doi: 10.1016/j.sjbs.2018.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Barsoum I.B., Smallwood C.A., Siemens D.R., Graham C.H. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665–674. doi: 10.1158/0008-5472.CAN-13-0992. [DOI] [PubMed] [Google Scholar]
  • 83.Doedens A.L., Stockmann C., Rubinstein M.P., Liao D., Zhang N., DeNardo D.G. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70(19):7465–7475. doi: 10.1158/0008-5472.CAN-10-1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Noman M.Z., Desantis G., Janji B., Hasmim M., Karray S., Dessen P. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211(5):781–790. doi: 10.1084/jem.20131916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chen T.C., Wu C.T., Wang C.P., Hsu W.L., Yang T.L., Lou P.J. Associations among pretreatment tumor necrosis and the expression of HIF-1α and PD-L1 in advanced oral squamous cell carcinoma and the prognostic impact thereof. Oral Oncol. 2015;51(11):1004–1010. doi: 10.1016/j.oraloncology.2015.08.011. [DOI] [PubMed] [Google Scholar]
  • 86.Lewis C.E., Pollard J.W. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66:605–612. doi: 10.1158/0008-5472.CAN-05-4005. [DOI] [PubMed] [Google Scholar]
  • 87.Melton D.W., McManus L.M., Gelfond J.A., Shireman P.K. Temporal phenotypic features distinguish polarized macrophages in vitro. Autoimmunity. 2015;48(3):161–176. doi: 10.3109/08916934.2015.1027816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mosser D.M., Edwards J.P. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(December (12)):958–969. doi: 10.1038/nri2448. Erratum in: Nat Rev Immunol.2010 Jun;10(6):460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Barros M.H., Hauck F., Dreyer J.H., Kempkes B., Niedobitek G. Macrophage polarisation: an immunohistochemical approach for identifying M1 and M2 macrophages. PLoS One. 2013;8(11) doi: 10.1371/journal.pone.0080908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sakakura K., Takahashi H., Kaira K., Toyoda M., Murata T., Ohnishi H. Relationship between tumor-associated macrophage subsets and CD47 expression in squamous cell carcinoma of the head and neck in the tumor microenvironment. Lab Invest. 2016;96(9):994–1003. doi: 10.1038/labinvest.2016.70. [DOI] [PubMed] [Google Scholar]
  • 91.Richards D.M., Hettinger J., Feuerer M. Monocytes and macrophages in cancer: development and functions. Cancer Microenviron. 2013;6:179–191. doi: 10.1007/s12307-012-0123-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Alves A.M., Diel L.F., Lamers M.L. Macrophages and prognosis of oral squamous cell carcinoma: a systematic review. J Oral Pathol Med. 2018;47(5):460–467. doi: 10.1111/jop.12643. Epub 2017 October 6. [DOI] [PubMed] [Google Scholar]
  • 93.Grotz T.E., Mansfield A.S., Jakub J.W., Markovic S.N. Regional lymphatic immunity in melanoma. Melanoma Res. 2012;22(1):9–18. doi: 10.1097/CMR.0b013e32834e1f33. [DOI] [PubMed] [Google Scholar]
  • 94.Wehrhan F., Büttner-Herold M., Hyckel P., Moebius P., Preidl R., Distel L. Increased malignancy of oral squamous cell carcinomas (oscc) is associated with macrophage polarization in regional lymph nodes—an immunohistochemical study. BMC Cancer. 2014;14:522. doi: 10.1186/1471-2407-14-522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wehrhan F., Büttner-Herold M., Distel L., Ries J., Moebius P., Preidl R. Galectin 3 expression in regional lymph nodes and lymph node metastases of oral squamous cell carcinomas. BMC Cancer. 2018;18(1):823. doi: 10.1186/s12885-018-4726-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kubota K., Moriyama M., Furukawa S., Rafiul H.A.S.M., Maruse Y., Jinno T. Nakamura S. CD163(+)CD204(+) tumor-associated macrophages contribute to T cell regulation via interleukin-10 and PD-L1 production in oral squamous cell carcinoma. Sci Rep. 2017;7(1):1755. doi: 10.1038/s41598-017-01661-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Zhang Y., Liu S., Qu D., Wang K., Zhang L., Jing X. Kif4A mediate the accumulation and reeducation of THP-1 derived macrophages via regulation of CCL2-CCR2 expression in crosstalking with OSCC. Sci Rep. 2017;7(1):2226. doi: 10.1038/s41598-017-02261-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhuang Z., Yoshizawa-Smith S., Glowacki A., Maltos K., Pacheco C., Shehabeldin M. Induction of M2 macrophages prevents bone loss in murine periodontitis models. J Dent Res. 2019;98(2):200–208. doi: 10.1177/0022034518805984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Qidwai T. Chemokine genetic polymorphism in human health and disease. Immunol Lett. 2016;176:128–138. doi: 10.1016/j.imlet.2016.05.018. [DOI] [PubMed] [Google Scholar]
  • 100.Yang L.L., Zhang M.J., Wu L., Mao L., Chen L., Yu G.T. LAIR-1 overexpression and correlation with advanced pathological grade and immune suppressive status in oral squamous cell carcinoma. Head Neck. 2019;41(4):1080–1086. doi: 10.1002/hed.25539. [DOI] [PubMed] [Google Scholar]
  • 101.Jiang C., Yuan F., Wang J., Wu L. Oral squamous cell carcinoma suppressed antitumor immunity through induction of PD-L1 expression on tumor-associated macrophages. Immunobiology. 2017;222(4):651–657. doi: 10.1016/j.imbio.2016.12.002. [DOI] [PubMed] [Google Scholar]
  • 102.Fujii N., Shomori K., Shiomi T., Nakabayashi M., Takeda C., Ryoke K. Cancer-associated fibroblasts and CD163-positive macrophages in oral squamous cell carcinoma: their clinicopathological and prognostic significance. J Oral Pathol Med. 2012;41(6):444–451. doi: 10.1111/j.1600-0714.2012.01127.x. [DOI] [PubMed] [Google Scholar]
  • 103.Takahashi H., Sakakura K., Kudo T., Toyoda M., Kaira K., Oyama T. Cancer-associated fibroblasts promote an immunosuppressive microenvironment through the induction and accumulation of protumoral macrophages. Oncotarget. 2017;8(5):8633–8647. doi: 10.18632/oncotarget.14374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Dinarello C.A. Interleukin-1α neutralisation in patients with cancer. Lancet Oncol. 2014;15(6):552–553. doi: 10.1016/S1470-2045(14)70164-0. [DOI] [PubMed] [Google Scholar]
  • 105.Coleman K.M., Gudjonsson J.E., Stecher M. Open-label trial of MABp1, a true human monoclonal antibody targeting interleukin 1α, for the treatment of psoriasis. JAMA Dermatol. 2015;151:555–556. doi: 10.1001/jamadermatol.2014.5391. [DOI] [PubMed] [Google Scholar]
  • 106.Germano S., Kennedy S., Rani S., Gleeson G., Clynes M., Doolan P. MAGE-D4B is a novel marker of poor prognosis and potential therapeutic target involved in breast cancer tumorigenesis. Int J Cancer. 2012;130(9):1991–2002. doi: 10.1002/ijc.26200. [DOI] [PubMed] [Google Scholar]
  • 107.Ito S., Kawano Y., Katakura H., Takenaka K., Adachi M., Sasaki M. Expression of MAGE-D4, a novel MAGE family antigen, is correlated with tumor-cell proliferation of non-small cell lung cancer. Lung Cancer. 2006;51(1):79–88. doi: 10.1016/j.lungcan.2005.08.012. [DOI] [PubMed] [Google Scholar]
  • 108.Montoro J.R., Mamede R.C., Neder Serafini L., Saggioro F.P., Figueiredo D.L., Silva W.A., Jr Expression of cancer-testis antigens MAGE-A4 and MAGE-C1 in oral squamous cell carcinoma. Head Neck. 2012;34(8):1123–1128. doi: 10.1002/hed.21880. [DOI] [PubMed] [Google Scholar]
  • 109.Xiao J., Chen H.S. Biological functions of melanoma-associated antigens. World J Gastroenterol. 2004;10(13):1849–1853. doi: 10.3748/wjg.v10.i13.1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Chong C.E., Lim K.P., Gan C.P., Marsh C.A., Zain R.B., Abraham M.T. Over-expression of MAGED4B increases cell migration and growth in oral squamous cell carcinoma and is associated with poor disease outcome. Cancer Lett. 2012;321(1):18–26. doi: 10.1016/j.canlet.2012.03.025. [DOI] [PubMed] [Google Scholar]
  • 111.Brisam M., Rauthe S., Hartmann S., Linz C., Brands R.C., Kübler A.C. Expression of MAGE-A1-A12 subgroups in the invasive tumor front and tumor center in oral squamous cell carcinoma. Oncol Rep. 2016;35(4):1979–1986. doi: 10.3892/or.2016.4600. [DOI] [PubMed] [Google Scholar]
  • 112.Weber M.M., Fottner C. Immune checkpoint inhibitors in the treatment of patients with neuroendocrine neoplasia. Oncol Res Treat. 2018;41(5):306–312. doi: 10.1159/000488996. [DOI] [PubMed] [Google Scholar]
  • 113.Smyth E., Thuss-Patience P.C. Immune checkpoint inhibition in gastro-oesophageal Cancer. Oncol Res Treat. 2018;41(5):272–280. doi: 10.1159/000489099. [DOI] [PubMed] [Google Scholar]
  • 114.Chen Y.M. Immune checkpoint inhibitors for nonsmall cell lung cancer treatment. J Chin Med Assoc. 2017;80(1):7–14. doi: 10.1016/j.jcma.2016.08.005. [DOI] [PubMed] [Google Scholar]
  • 115.Ibrahim R., Stewart R., Shalabi A. PD-L1 blockade for cancer treatment: MEDI4736. Semin Oncol. 2015;42(June (3)):474–483. doi: 10.1053/j.seminoncol.2015.02.007. [DOI] [PubMed] [Google Scholar]
  • 116.Taube J.M., Klein A., Brahmer J.R., Xu H., Pan X., Kim J.H. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014;20(19):5064–5074. doi: 10.1158/1078-0432.CCR-13-3271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Herbst R.S., Soria J.C., Kowanetz M., Fine G.D., Hamid O., Gordon M.S. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–567. doi: 10.1038/nature14011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Wang J., Xie T., Wang B., William W.N., Jr, Heymach J.V., El-Naggar A.K. PD-1 blockade prevents the development and progression of carcinogen-induced oral premalignant lesions. Cancer Prev Res (Phila) 2017;10(12):684–693. doi: 10.1158/1940-6207.CAPR-17-0108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Li L., Lu S., Liang X., Cao B., Wang S., Jiang J. γδTDEs: an efficient delivery system for miR-138 with anti-tumoral and immunostimulatory roles on oral squamous cell carcinoma. Mol Ther Nucleic Acids. 2019;14:101–113. doi: 10.1016/j.omtn.2018.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Maeda A., Digifico E., Andon F.T., Mantovani A., Allavena P. Poly(I:C) stimulation is superior than Imiquimod to induce the antitumoral functional profile of tumor-conditioned macrophages. Eur J Immunol. 2019;49(5):801–811. doi: 10.1002/eji.201847888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yuan H., He J., Zhang G., Zhang D., Kong X., Chen F. Osteoclast stimulatory transmembrane protein induces a phenotypic switch in macrophage polarization suppressing an M1 pro-inflammatory state. Acta Biochim Biophys Sin (Shanghai) 2017;49(10):935–944. doi: 10.1093/abbs/gmx092. [DOI] [PubMed] [Google Scholar]
  • 122.Zhu W., Xu R., Du J., Fu Y., Li S., Zhang P. Zoledronic acid promotes TLR-4-mediated M1 macrophage polarization in bisphosphonate-related osteonecrosis of the jaw. FASEB J. 2019 doi: 10.1096/fj.201801791RR. January 9. [DOI] [PubMed] [Google Scholar]
  • 123.Kim R. Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J Transl Med. 2018;16(1):8. doi: 10.1186/s12967-018-1389-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Colucci D.G., Puig N.R., Hernandez Pando R. Influence of anaesthetic drugs on immune response: from inflammation to immunosuppression. Oa Anaesth. 2013;1(December 30 (3)):21. [Google Scholar]

Articles from The Japanese Dental Science Review are provided here courtesy of Elsevier

RESOURCES