Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jul 1.
Published in final edited form as: Oral Oncol. 2016 May 20;58:59–70. doi: 10.1016/j.oraloncology.2016.05.002

Overcoming barriers to effective immunotherapy: MDSCs, TAMs, and Tregs as mediators of the immunosuppressive microenvironment in head and neck cancer

Ruth J Davis 1, Carter Van Waes 1, Clint T Allen 1,2
PMCID: PMC4912416  NIHMSID: NIHMS786891  PMID: 27215705

Abstract

A significant subset of head and neck cancers display a T-cell inflamed phenotype, suggesting that patients with these tumors should respond to therapeutic approaches aimed at strengthening anti-tumor immune responses. A major barrier to the development of an effective anti-tumor immune response, at baseline or in response to immunotherapy, is the development of an immunosuppressive tumor microenvironment. Several well described mechanisms of effector immune cell suppression in the head and neck cancer microenvironment are discussed here, along with updates on current trials designed to translate what we have learned from pre-clinical and correlative clinical studies into improved responses in patients with head and neck cancer following immune activating therapies.

Introduction

The Role of Immunity in Head and Neck Cancers

Advances in understanding the role of the immune system in preventing the development and progression of clinically significant cancers have encouraged interest in therapeutic strategies targeting immunity. Head and neck squamous cell carcinoma (HNSCC), with its poor prognosis despite refinements in surgical and chemoradiation protocols, is no exception. The Cancer Genome Atlas (TCGA) has clarified the common genetic alterations contributing to development and progression of HNSCC, and the rational design of agents targeting specific oncogenic pathways in HNSCC remains a valuable approach that can be effective in subsets of patients [1]. However, the heterogeneity of human tumors, ability of plastic cancer cells to adaptively resist single agent small molecule inhibitors, and toxicity commonly observed with combination small molecule inhibitors represent significant challenges [24]. Immune-based therapies are less likely to depend on common genetic alterations and carry the potential to generate systemic, durable anti-tumor responses. Compared to many other malignancies, HNSCC has a high genetic mutation rate and generates immunogenic tumors with robust immune cell infiltration [58]. Accordingly, a significant subset of patients HNSCC patients objectively respond to immunotherapies such as antibody-based checkpoint inhibition [9]. For these reasons and others, immune-based therapeutic strategies dominate the landscape of new clinical trials in cancer treatment [10].

Key mediators of effective anti-tumor immune responses include tumor-infiltrating lymphocytes (TILs) and natural killer (NK) cells. Activated TILs are cytotoxic to target cells in an MHC-restricted, antigen-specific fashion through well-defined mechanisms of cytotoxicity [11,12]. Alternatively, NK cells, broadly considered to be part of the innate immune response, recognize tumor cells through non-specific mechanisms including detection of low cell surface MHC [13]. Increased TILs and tumor-infiltrating NK cells correlate with better response to standard therapies and, accordingly, prognosis and survival [14,15].

Cancer development and progression is predicated upon evasion of immune recognition and destruction. Mechanisms of immune escape can be divided into lack of initial activation of an anti-tumor immune response and/or suppression of an activated response. Although both mechanisms are likely crucial to tumor development and progression, the majority of HNSCCs demonstrate measurable infiltration of effector immune cells suggesting the presence of immune suppression within the tumor microenvironment (TME) [6,8,16]. Perhaps even more critically, these same mechanisms of immunosuppression that allow an upper aerodigestive tract malignancy to develop and progress are likely the same that confer resistance to immune activating treatments. Understanding how to modulate the TME will be critical as we aim to enhance patient responses to such treatments. This review will focus on known mechanisms of immunosuppression in the HNSCC microenvironment, with particular attention to myeloid-derived suppressor cells (MDSCs), polarized macrophages, regulatory T cells (Tregs), and work being done to translate pre-clinical knowledge into clinical advances.

Discussion

Mechanisms of Immunosuppression in the Tumor Microenvironment

Many different cell types and functional states contribute to the overall immunosuppressive or immunopermissive status of the TME, and this can vary between patients and even regionally within one tumor [17]. The ability to characterize the overall immune status of large cohorts of tumors has been aided by genomic sequencing and array profiling techniques [1,6], but is still handicapped by tumor sampling and difficulties analyzing representative tissue from a heterogeneous source. In its most basic form, immunosuppression within the TME is mediated directly by HNSCC tumor cells or indirectly by the stroma or through recruitment and polarization of host cells [10,16,18]. All of these factors are important, and ultimately the immunogenicity of an individual HNSCC tumor (and subsequent response to immune-activating therapies) is determined by the balance between recruitment and activation of innate and antigen-specific effector immune cells, and immunosuppressive forces levied by tumor, stromal and other infiltrating immune cells.

Direct Tumor Effects

The physical characteristics of tumors themselves represent a barrier to the infiltration of immune cells. Rapid cellular growth during tumor development leads to formation of abnormal vasculature and lymphatics, which are inherently leaky and cause elevated interstitial pressure within the tumor that physically blocks immune cell infiltration [19]. This abnormal vasculature also contributes to a hypoxic TME, which induces secretion of T-cell inhibiting compounds galectin-1 and adenosine [19,20].

While immune cells overcome these barriers and infiltrate into tumor cell nests in many HNSCC tumors, unfortunately HNSCC cells utilize many direct and indirect mechanisms of immune suppression and tumor cells with this capacity are likely selected for during tumorigenesis [21]. HNSCC tumor cells can directly express immunosuppressive cytokines TGF-β, IL-6 and IL-10 that suppress T-cell proliferation and cytolytic function [22,23]. In addition to direct release of immunosuppressive cytokines, tumor cell metabolism can deplete nutrients critical to effector immune cell function [24,25]. Overexpression of enzymes within cancer cells such as indoleamine 2,3-dioxygenase (IDO1) deplete local tryptophan pools, profoundly inhibiting T-lymphocyte function [26].

Cancer cells also express surface ligands that bind and inhibit effector T-cells [27]. Known as checkpoints, T-cells express positive and negative co-stimulatory receptors on their surface that normally function to regulate immune responses and prevent autoimmunity. Well known checkpoints expressed on activated T-cells include programmed death 1 (PD1) and cytotoxic T-lymphocyte protein 4 (CTLA4) [28]. Checkpoint ligands, including PD-Ligand 1 (PD-L1) and CD80, are often expressed on HNSCC cells either constitutively downstream of oncogenic signaling pathways or in response to interferon (IFN) present within the TME [29,30]. Induced expression of PD-L1 in response to IFN from activated local T-cells and NK cells is termed adaptive resistance and represents a major barrier to effective immune cell function in an otherwise T-cell inflamed TME [28,31].

Role of Host Cells

In addition to direct mechanisms of immunosuppression detailed above, HNSCC cells also secrete chemokines that recruit hematopoietic cells to the TME [32]. These include cytotoxic immune cells required for an effective immune response, but also include many types of immunosuppressive immune cells. Immature or naïve immune cells that are highly plastic can be polarized into an immunosuppressive phenotype by the cytokine milieu within the TME after their recruitment [10,16]. Well-characterized cell subsets are discussed here.

Myeloid derived suppressor cells

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of immature myeloid cells that were first characterized in humans as a population of CD34+ suppressor cells in patients with HNSCC [33]. Around the same time, a mouse immunosuppressive myeloid cell equivalent was characterized in syngeneic models of HNSCC [34,35]. In healthy humans these immature cells rapidly differentiate into mature macrophages, granulocytes, or dendritic cells and only make up ~0.5% of peripheral blood mononuclear cells (PBMCs) [36]. However, in cancer patients this differentiation is partially arrested, resulting in expansion of the immature population [32]. The presence of expanded MDSCs peripherally and within the TME is associated with metastasis and recurrence after definitive treatment in HNSCC patients [33,37]. Manipulation of MDSCs within the TME alters effector T-cell function and responses to anti-cancer therapies, making MDSCs a major focus of research [33,38,39].

Mechanisms of MDSC Recruitment

MDSCs are characterized as CD11b+CD14+CD33+HLA-DR-in humans [32]. In mice, two MDSC subtypes have been identified: CD11b+Ly6G+Ly6Cint granulocytic MDSCs (gMDSCs) and CD11b+Ly6GlowLy6Chi monocytic MDSCs (mMDSCs). Similar subsets of MDSCs have been characterized in humans. Both subtypes suppress T-cell activity, and the relative role of gMDSCs and mMDSCs appears to be model dependent.

MDSCs are recruited into the HNSCC TME by a chemotactic gradient involving GM-CSF, MCP-1, CXCL1, IL-8, and CSF1 released from the TME (Figure 1) [38,4042]. Murine mMDSCs often express the chemokine receptor CCR2, and melanoma studies in transgenic mice have demonstrated recruitment of this population into the TME with subsequent T-cell suppression in response to tumor produced GM-CSF [41]. gMDSCs express chemokine receptors CXCR2 and CSFR1, for which CXCL1, CXCL8 (IL-8 in humans) and CSF1 are the primary ligands. Indeed, murine and human HNSCC often significantly express CXCL1 and IL-8 [43]. Studies using CXCR2-deficient mice or CXCR2 blocking antibodies have demonstrated decreased gMDSC accumulation with subsequent decreased tumorigenesis, decreased primary tumor growth rates and enhanced responses to therapy [38,40,44], strongly pointing to MDSCs as a major barrier to effective immunotherapy and immune clearance of tumor cells.

Figure 1. Myeloid Cell Differentiation and Recruitment.

Figure 1

Myeloid-derived suppressor cells (MDSCs) accumulate under the influence of factors including GM-CSF, IL-6, IL-10, COX2, IL-1β, IDO, and VEGF, and are recruited to the tumor microenvironment by secreted CXCL1, IL-8, GM-CSF and CSF1. gMDSCs are terminally differentiated, while mMDSCs can further differentiate into macrophages and dendritic cells. Macrophages can also be recruited to the tumor microenvironment by CSF1.

While peripheral MDSCs express chemokine receptors and can be recruited into the HNSCC TME, some evidence exists that PBMCs can be recruited and converted into MDSCs by the local TME cytokine milieu. G-CSF, M-CSF, IL-10, prostaglandins, TNF-α and VEGF have all been shown to induce MDSCs from PBMCs [32,45,46]. Cytokines expressed by HNSCC cell lines can directly contribute to the generation of MDSCs from PBMCs in vitro, with IL-6 and GM-CSF playing dominant roles [46]. These inflammatory cytokines and chemokines are constitutively expressed by many HNSCC cell lines and tumors secondary to upstream genetic alterations, forming a link between dysregulated oncogenic signaling and development of an immunosuppressive TME [35,47].

Mechanisms of MDSC immunosuppression

Classic mechanisms of T-cell suppression by MDSCs that have been demonstrated in many tumor models include production of arginase 1 (Arg-1), nitric oxide synthase (iNOS), and reactive oxygen species (ROS) (Figure 2) [32]. L-arginine is a common substrate for Arg-1 and iNOS. Upregulation of these enzymes depletes L-arginine from the TME, which directly inhibits T-cells. Expression of Arg-1 depends on COX2-mediated production of prostagladin E2 [48]. iNOS converts L-arginine into nitric oxide, which is subsequently converted into peroxynitrite. This reactive nitrogen species potently suppresses T-cell function and correlates with a poor prognosis in a variety of cancers [32,45].

Figure 2. Immunosuppressive Mechanisms of MDSCs.

Figure 2

Under the influence of transcription factors STAT1, STAT3, STAT6 and HIF1α, MDSCs suppress TILs through a variety of mechanisms. Arg-1 inhibits TILs through depletion of arginine and activates Tregs. iNOS inhibits TILs through production of reactive oxygen and nitrogen species in addition to depletion of environmental arginine. MDSCs also secrete immunosuppressive cytokines IL-6 and IL-10, and express the immune checkpoint ligand PD-L1. Intracellular IDO expression has also been proposed as a mechanism of TIL inhibition through depletion of tryptophan and production of kynurenine metabolites.

Recently, additional metabolic mechanisms of MDSC-mediated immunosuppression have been identified. Similar to tumor cells themselves, altered tryptophan metabolism through IDO expression can deplete local tryptophan and suppress T-cell function [49]. Further, IDO catalyzes conversion of tryptophan into immunosuppressive kynurenine metabolites. MDSCs also deplete environmental cysteine, which T-cells are unable to synthesize for themselves [50]. The MDSC-mediated depletion of metabolites required for effective T-cell function represents a formidable barrier to anti-tumor immunity in a hostile TME characterized by intense competition for metabolic substrates.

Signal transducer and activator of transcription (STAT) family members are key transcription factors responsible for development of an MDSC phenotype in myeloid populations [32]. Increased STAT3 activation has been observed in MDSCs of tumor-bearing compared to naïve mice, and this activation has been associated with increased expression of pro-growth/survival genes Myc, Bcl-xL, and cyclin D1 [32]. Upregulation of MDSC Arg-1 is largely dependent upon STAT1, STAT3, and STAT6, whereas IDO has been linked to STAT3 and iNOS to STAT1 [32,49]. STAT activation within MDSCs is influenced by TME cytokines including IFNγ, IL-4, -6 and -13 as well as TGFβ [45]. In HNSCC patients, MDSCs isolated from the blood, lymph nodes, and HNSCC tumors have been shown to suppress T-cell proliferation at least through STAT3-dependent Arg-1 activity [51].

The hypoxic TME influences MDSC function through the transcription factor hypoxia-inducible factor-1α (HIF-1α) [52]. HIF-1α induces PD-L1 expression on MDSCs and other immune cells within the TME. Interestingly, antibody blockade of PD-L1 function following HIF-1α induced overexpression appears to reduce MDSC production of the inflammatory and immunosuppressive cytokines IL-6 and 10, suggesting a link between hypoxia and multiple immunosuppressive mechanisms of MDSCs. Peripheral MDSCs from HNSCC patients also suppress T-cell function through surface expression of PD-L1 and the release of TGFβ [53], suggesting a role for PD-L1 in MDSC-mediated T-cell suppression outside of the hypoxic tumor microenvironment as well.

MDSCs also appear to be capable of functionally suppressing effector T-cells in an antigen-specific fashion. MDSC presentation of peptide to antigen-specific T-cells results in nitration of T-cell surface molecules, leading to antigen-specific T-cell receptor (TCR) dysfunction and subsequent unresponsiveness to the same antigen presented by professional antigen presenting cells (APCs) [54]. This suggests that the presence of MDSCs in the TME of an otherwise antigenic tumor may result in silencing of antigen-specific T-cells. Another significant immunosuppressive mechanism of MDSCs is the activation of CD4 regulatory T cells (Tregs). Tregs are a FoxP3+CD25+ subset of CD4 T-cells that normally maintain self-tolerance, but can be dysregulated in the TME to cause local immunosuppression [55].

The link between MDSCs and Tregs has been established in many different animal models [56], and has been confirmed in MDSCs derived from cancer patients [57]. In various cancer models, MDSC-mediated induction and activation of Tregs depends upon IL-10, IFNγ, CD40, and arginase expression and may occur following uptake of tumor antigen and activation of antigen-specific Tregs [56,58,59]. Through Treg activation and induced TCR dysfunction, MDSCs suppress antigen-specific adaptive immune responses. Clearly, the presence of MDSCs in the TME potently suppresses anti-tumor immunity and alters the inflammatory balance in favor of immune escape and tumor progression.

Targeting MDSCs in HNSCC

Given the above evidence, suppression of either MDSC function or recruitment has become a major research focus for many cancer types including HNSCC. A variety of approaches including forced differentiation, small molecule inhibitors and chemotaxis blockade are under investigation in various stages of development (Figure 3).

Figure 3. Therapeutic Targeting of MDSCs.

Figure 3

CXCR2 and CSFR1 antibodies inhibit chemotaxis of MDSCs to the tumor microenvironment. Vitamin D3 reduces MDSC numbers by promoting differentiation into macrophages and dendritic cells. PDE5 inhibitors increase intracellular cGMP, thereby inhibiting the immunosuppressive enzymes iNOS and Arg-1. Small molecule inhibitors of IDO, iNOS, Arg-1, and STAT3 have also been developed. Depleting antibodies targeted against Gr1 and Ly6G surface markers reduce MDSC numbers.

Vitamin D3 Analogs

As MDSCs represent an immature myeloid cell phenotype, initial efforts focused on inducing differentiation of these immature cells into mature, less immunosuppressive phenotypes. 1,25-dihydroxyvitamin D3 induces differentiation of peripheral blood CD34+ MDSCs isolated from HNSCC patients into antigen-presenting dendritic cells [60]. Although no objective responses were observed, an initial pilot of 1,25-hydroxyvitamin D3 in patients with advanced HNSCC demonstrated reduction in peripheral MDSCs and increased plasma concentrations of IL-12 and IFN-γ [61]. Further study revealed decreased MDSC accumulation, increased mature dendritic cell, CD4 and CD8 T-cell and enhanced T-cell CD69 expression in the TME of HNSCC patients treated with neoadjuvant 1,25-dihydroxyvitamin D3 before surgical extirpation [62]. Given these promising immune correlates and low toxicity profile, a clinical trial of 1,25-hydroxyvitamin D3 in combination with the COX2 inhibitor celecoxib in surgically resectable oral cavity HNSCC has recently been completed, with results pending publication (NCT00953849).

Small Molecule Inhibitors

As two well-known mediators of MDSC immunosuppression that act through L-arginine depletion, Arg-1 and iNOS presented promising targets. Although inhibition of Arg-1 with nor-NOHA and iNOS with L-NMMA mechanistically reduced the suppressive effect of murine and human tumor-derived MDSCs, these inhibitors are limited by their toxicity in humans [63].

Alternatively, identification of IDO as a mechanism of MDSC immunosuppression has led to development of IDO inhibitors capable of inhibiting MDSC function in vitro [49]. A number of phase I/II clinical trials using IDO inhibitors alone (NCT02048709) or in combination with other immune-based therapies (NCT02327078, NCT02178722, NCT02559492) are currently underway in patients with advanced solid tumors. See table 1 for a list of active immunotherapy-based clinical trials discussed in this manuscript. For an exhaustive discussion of immunotherapy trials in currently enrolling HNSCC patients, please refer to a recent review by Allen, et al. [10].

Table 1.

Current Immunotherapy Trials in HNSCC Targeting the Immunosuppressive Tumor Microenvironment

Targets Treatments Phase Clinical Trial ID Patient Eligibility
IDO1 GDC-0919 I NCT02048709 Recurrent/refractory solid tumors
IDO1
PD-1
Epacadostat +
Nivolumab
I/II NCT02327078 Recurrent/metastatic solid
tumors including HNSCC
IDO1
PD-1
INCB024360
(Epacadostat) +
MK-3475
(Pembrolizumab)
I/II NCT02178722 Recurrent/metastatic solid
tumors including HNSCC
IDO1
JAK1
INCB024360
(Epacadostat) +
INCB039110
Ib NCT02559492 Advanced/metastatic solid
tumors including HNSCC
PD-L1
STAT3
CXCR2
MEDI4736 +
AZD9150 or
AZD5069
Ib/II NCT02499328 Recurrent/metastatic HNSCC
PDE5 Tadalafil II NCT01697800 Untreated nonmetastatic
HNSCC
PDE5
Tumor Mucin 1
(MUC1)
Tadalafil
Anti-MUC1 Vaccine
I/II NCT02544880 Resectable HNSCC
CSF1R
PD-1
PLX3397 +
MK-3475
(Pembrolizumab)
I/IIa NCT02452424 Recurrent/metastatic solid
tumors including HNSCC
CSF1R
PD-1
FPA008 +/−
Nivolumab
Ia/Ib NCT02526017 Advanced solid tumors including
HNSCC
CCR4
PD-L1
CTLA-4
KW-0761
(Mogamulizumab)
+ MEDI4736 or
Tremelimumab
I NCT02301130 Advanced solid tumors including
HNSCC
CCR4
PD-1
KW-0761
(Mogamulizumab)
+ Nivolumab
I NCT02705105 Advanced/metastatic solid
tumors
CCR4
4-1BB
KW-0761
(Mogamulizumab)
+ PF-05082566
Ib NCT02444793 Advanced/metastatic solid
tumors
CCR4 KW-0761
(Mogamulizumab)
I/II NCT02281409 Advanced/metastatic solid
tumors
OX40
4-1BB
PF-04518600 +/−
PF-05082566
I NCT02315066 Advanced/metastatic solid
tumors including HNSCC
OX40 MEDI6469 Ib NCT02274155 Resectable HNSCC
GITR TRX518 I NCT01239134 Advanced/metastatic melanoma
or solid tumors
GITR TRX518 I NCT02628574 Advanced solid tumors
GITR MEDI1873 I NCT02583165 Advanced solid tumors

As a transcriptional regulator of Arg-1 and IDO expression in MDSCs, STAT3 represents another logical target. A number of STAT3 inhibitors are in development and have successfully reduced MDSC function through Arg-1 and IDO inhibition in preclinical experiments [49,51]. Success in early phase trials of the STAT3 inhibitors OPB-51602 and OPB-31121 has been limited by toxicities, including early termination of one trial (NCT02058017) due to intolerable metabolic acidosis [64]. Recent development of the antisense oligonucleotide STAT3 inhibitor ISIS 481464 (AZD9150) has shown promise in initial clinical trials in lymphoma [65]. Currently, a phase I/II trial of PD-L1 blocking antibody in combination with AZD9150 is recruiting patients with recurrent/metastatic HNSCC (NCT02499328).

Targeted Antibodies

Initial pre-clinical work utilized anti-Ly6G or anti-Ly6C antibodies or depleted MDSCs systematically in mice, and this has led to promising anti-tumor and immune enhancing effects in multiple cancer models [66,67]. Translationally, most MDSCs express the chemokine receptor CXCR2. CXCR2 knockout mice demonstrate substantially reduced myeloid recruitment into the TME, and the majority of HNSCCs express significant levels of the CXCR2 ligand CXCL1. Thus, targeted inhibition of CXCR2 represents a rational therapeutic strategy. Monoclonal antibody blockade of CXCR2 results in reduced recruitment of MDSCs into the TME and primary tumor growth suppression [38,44]. Mechanistically, this may be due to increased TIL activation[38], removal of MDSC-liberated proinflammatory cytokines and growth factors[44], or both. More recent work has focused on the combination of CXCR2 blocking antibodies with immune activating therapies, such as checkpoint inhibitors. Indeed, eliminating CXCR2+ MDSCs from the TME enhances responses observed with PD1 targeting immunotherapy in different animal models [38]. A number of small molecule inhibitors of CXCR2 are also in development [68]. A clinical trial of an anti-PD-L1 antibody combined with a STAT3 inhibitor or anti-CXCR2 antibody for patients with metastatic HNSCC (NCT02499328) is underway.

Phosphodiesterase 5 Inhibitors

Phosphodiesterase 5 (PDE5) inhibitors such as sildenafil and tadalafil are used to treat erectile dysfunction and pulmonary hypertension. In addition to mediating these vasodilatory effects, PDE5 inhibition suppresses production of Arg-1 and iNOS, leading to functional inhibition of MDSCs [39]. In several syngeneic murine cancer models, sildenafil treatment slowed tumor growth, increased tumor infiltration with CD8+ T-cells, upregulated T-cell activation markers CD69 and CD25, and decreased Treg proliferation [37,39,58]. MDSCs from sildenafil-treated mice demonstrated decreased ability to suppress T-cell proliferation [39]. In addition, peripheral T-cells from HNSCC patients demonstrated recovery of proliferative capacity in response to ex vivo treatment with sildenafil.

Based on this promising pre-clinical data, tadalafil has been tested in two randomized placebo-controlled phase II trials in HNSCC [37,69]. In both studies, neoadjuvant tadalafil prior to definitive therapy was well tolerated and enhanced objective markers of anti-tumor immunity. Weed et al. observed a significant reduction in the numbers of MDSCs and Tregs in peripheral blood and the TME of treated patients compared to placebo [37]. The proliferative capacity of circulating CD8+ T-cells also improved following tadalafil treatment.

Califano et al conducted a parallel study comparing high-dose tadalafil to placebo, in which a reduction in peripheral MDSC numbers and an increase in ex vivo T-cell proliferation were also observed [69]. Consistent with previous results, MDSC expression of Arg-1 and iNOS were also reduced. This MDSC suppression correlated with enhanced anti-tumor immunity. Interestingly, evidence of enhanced CD4 and CD8 T-cell activation in response to tumor antigen was observed. Currently, tadalafil is in two phase II trials in HNSCC patients as an adjuvant to standard of care and immune activating treatments (NCT01697800 & NCT02544880).

Tumor Associated Macrophages

Mature macrophages, once recruited into the TME, exhibit high plasticity and appear to be heavily influenced by the TME cytokine milieu. Tumor-associated macrophages (TAMs) develop into one of two phenotypes: tumor limiting M1 or tumor enhancing M2. M2 TAMs support tumor growth through secretion of growth promoting cytokines and local immunosuppression, whereas M1 macrophages possess tumoricidal characteristics and can directly limit primary tumor growth [70,71]. Although the M1/M2 paradigm represents extreme phenotypes of a large spectrum of macrophage functions, in general the macrophage infiltrate in the HNSCC TME is primarily M2 in composition [72]. High levels of M2 TAM infiltrate correlate with metastasis and poor prognosis in HNSCC patients [8,72,73].

TAMs can be recruited to the tumor from the bone marrow, or arise from mMDSCs that differentiate into macrophages within the TME (Figure 1) [74]. Macrophage recruitment to the tumor is largely dependent upon colony stimulating factor 1 (CSF1) and monocyte chemotactic protein 1 (MCP-1) signaling with contributions from GM-CSF, VEGF and other chemokines [75]. Hypoxia appears to be a major driver of the induction of signals that recruit macrophages.

Mechanisms of TAM Immunosuppression

TAMs functionally contribute to the immunosuppressive TME through secretion of enzymes, soluble factors, and expression of surface molecules (Figure 4). Like MDSCs, M2 TAMs also express Arg-1 and iNOS, leading to depletion of L-arginine and metabolic suppression of T-cell function. Also like MDSCs, TAMs express surface PD-L1 and tend to be polarized toward an immunosuppressive M2 phenotype under hypoxic conditions [52,75]. In a recent analysis of pools of PD-L1 in the TME of syngeneic murine oral cavity HNSCC, macrophages represented the population of cells within the tumor with the highest intensity of PD-L1 staining [76]. TAMs also express the checkpoint ligands CD80/86 (B7-1/2) that inhibit TILs through binding CTLA4 [77]. In some cancer models, surface expression of specific MHC class I proteins can activate or suppress effector immune cells, with macrophage expression of less common HLA-G and E demonstrating ability to suppress T and NK cell function [78]. TAMs also secrete immunosuppressive cytokines IL-1β, IL-6, IL-10, and TGFβ [74]. IL-10 inhibits effector T-cell function and activates Tregs [79]. Further establishing the relationship between TAMs and other regulatory immune cells, TAMs (along with tumor cells, themselves) recruit Tregs to the TME through chemokines CCL22, CCL20, and CCL5 [8082].

Figure 4. Tumor-Associated Macrophages.

Figure 4

M2 macrophages inhibit T-cells through surface expression of HLA-G, CD80/86, and PDL1/PDL2. Like MDSCs macrophages secrete Arg1, which inhibits TILs through depletion of arginine. Macrophages also secrete immunosuppressive cytokines IL-1β, IL-6, IL-10 and TGFβ which inhibit TILs and activate Tregs. Surface expression of HLA-E also may inhibit NK cells in some cancer models. The central approach to targeting TAMs has been through the development of CSFR1 antagonists and antibodies.

Targeting TAMs in HNSCC

Chemotaxis blockade

A main approach to targeting TAMs in cancer therapy has focused on inhibiting their recruitment into the TME. mMDSCs share a dependence on CSF1/CSF1R for recruitment, so CSF1R inhibition may target both mMDSCs and TAMs. Studies of anti-CSF1R antibodies and small molecule inhibitors of CSF1R in a variety of cancer models have demonstrated depletion of TAMs from the TME and reprogramming of the remaining macrophages from the M2 pro-tumor to the M1 anti-tumor phenotype [42,83,84]. Although blockade of CSF1/CSF1R signaling resulted in only modest growth suppression in a murine cancer model, combination with anti-PD1 and anti-CTLA4 therapy resulted in regression of established tumors [42]. Anti-CSF1R antibodies have been used to deplete TAMs in patients with a variety of cancers, including one with advanced HNSCC [85]. Two trials combining anti-CSF1R antibody with PD-1 checkpoint blockade are currently recruiting patients with advanced solid tumors (NCT02452424 and NCT02526017). Antibody or tyrosine kinase inhibitor-mediated blockade of other chemokine systems including VEGF receptor and CXCL12 can also reduce accumulation of TAMs [75,83,86].

Reversing TAM polarization

Another attractive approach to targeting TAMs is inducing polarization away from an immunosuppressive M2 toward a cytolytic M1 phenotype. Not only does this approach decrease tumor promoting M2 TAMs, but increases anti-tumor M1 TAM activity. Successful approaches in animal models have included toll-like receptor 3 ligation with Poly(I:C) [87], nanoparticle-mediated delivery of hydrazinocurcumin [88], zoledronic acid treatment [89], and deposition of histidine-rich glycoproteins within the TME [90]. Recent evidence suggests that type I IFN signaling through STING agonists can also re-educate M2 TAMs into an M1 phenotype [91]. In mouse models, STING agonists improve responses to radiation therapy by enhancing the adaptive immune response against the tumor antigens released by radiation [92]. These approaches are in various stages of development and have great capacity to serve as adjuvants to standard and immune therapies moving forward.

Regulatory T-cells

Tregs represent the third major cell type that contributes to the immunosuppressive TME in HNSCC. These cells are a subset of CD4 T-cells that express the transcription factor FoxP3 and commonly express CD25, CTLA4, glucocorticoid-induced TNF receptor (GITR) and OX40 (CD134) on their cell surface [55]. Studies have shown mixed results with regards to the prognostic value of tumor-infiltrating Tregs in HNSCC. In some studies, increased peripheral and tumor-infiltrating Tregs were associated with a negative prognosis in HNSCC patients [37,93]. However, Badoual, et. al. observed the opposite, with Treg infiltration correlating with favorable locoregional control [94]. These authors hypothesized that this unexpected result may be due to the ability of Tregs to inhibit tumor-promoting inflammation as well as anti-tumor immunity. Interestingly, greater numbers of circulating Tregs were observed in HNSCC patients with complete response to treatment compared to untreated patients with active disease, indicating that anti-cancer therapies may increase Treg expansion and provide a rationale for therapeutically targeting them to prevent recurrence [95].

Mechanisms of Treg recruitment

Tregs can be broadly subclassified as either natural or inducible Tregs. Natural Tregs develop as CD4+CD25+ Tregs in the thymus and are subsequently recruited into the TME [82,96]. Inducible Tregs, on the other hand, are undifferentiated CD4+ T-cells that are polarized into CD4+CD25+ Tregs by the HNSCC cytokine profile [97]. Tumor-infiltrating Tregs demonstrate more potent immunosuppression than those in the periphery, suggesting that the HNSCC TME influences the suppressive activity of these cells [98]. Evidence supports recruitment of Tregs to the TME through tumor-derived CCL22 binding the chemokine receptor CCR4 on the surface of Tregs [55,82]. In melanoma patients, tumor-infiltrating FOXP3+ T-cells were found to primarily express CCR4, whereas peripheral Tregs lacked CCR4, suggesting a critical role for this receptor in Treg trafficking [99]. Additional signals implicated in Treg recruitment to the TME include CCL28-CCL28R and CXCL10-CXCR3 interactions [100,101].

FOXP3-T-cells can be converted into FOXP3+ Tregs within the TME in the presence of IL-10, TGF-β and retinoic acid [56,102,103]. CD80 binding of CTLA4 on the surface of effector T-cells also induces differentiation into TGF-β+ Tregs in an IL-10-dependent manner [104]. On the transcription factor level, STAT3 appears to be necessary for FoxP3 expression and an immunosuppressive phenotype [105]. Whether they are recruited to the TME or induced from undifferentiated CD4 T-cells already present in the TME, Tregs in the TME expand following recognition of tumor-associated self antigens in the presence of IL-2 [55,106]. Further, mounting evidence suggests that Tregs mediate their immunosuppressive effects in an antigen-specific manner [107,108]. These concepts have critical implications for rational design of immunotherapy approaches involving Treg manipulation.

Mechanisms of Treg immunosuppression

Tregs derived from HNSCC patients suppress effector immune cell function, at least in part, via the production of IL-10 and TGF-β [109,110]. Studies have also demonstrated the ability of tumor-infiltrating Tregs to kill effector NK and CD8+ T-cells in a granzyme B- and perforin-dependent manner [111]. In addition, tumor-infiltrating Tregs in HNSCC patients express the cell surface molecules CTLA4 and CD39 as well as surface bound TGF-β1 [98]. Conditional knockout of CTLA4 in Tregs impaired their suppressive function and protected from tumor development [112]. CTLA4 downregulates CD80/CD86 expression by APCs and thereby limits CD28 binding and costimulation of naïve T-cells. CD39 (ENTPD1) on the surface of Tregs catalyzes degradation of ATP and ADP into AMP, which is subsequently degraded into immunosuppressive adenosine by CD73 [113].

Targeting Tregs in HNSCC

Strategies aimed at targeting Tregs in the TME have focused on surface markers including CD25, CCR4, CTLA4, OX40 and GITR (Figure 5).

Figure 5. Therapeutic Targeting of Regulatory T-Cells (Tregs).

Figure 5

In addition to secreting immunosuppressive cytokines IL-10 and TGFβ, Tregs inhibit TILs through surface expression of CTLA4 and CD39. Antibodies against surface molecules CTLA4, CCR4, and CD25 have all been shown to deplete Tregs in various cancer models. Agonist antibodies against GITR and OX40 stimulate effector T-cells in addition to inhibiting Treg function.

Anti-CD25 Monoclonal Antibodies

CD25 is the α chain of the IL-2 receptor required for effector function. It is constitutively expressed on the surface of many Tregs, making it a target for monoclonal antibody development. This strategy has shown promise in preclinical studies in a variety of cancer types [114116], but clinical studies have shown mixed results. In a trial of metastatic melanoma patients treated with anti-CD25 mAb in combination with a dendritic cell vaccine, activated effector T-cells expressing CD25 were depleted in addition to the targeted Tregs, potentially contributing to a lack of observed clinical responses [114]. Conversely, a recent clinical trial of metastatic breast cancer patients treated with anti-CD25 mAb in combination with peptide vaccination demonstrated depletion of Tregs and improved peptide-specific CD4/CD8 T-cell responses [115]. Clearly more work is needed to understand the impact of therapeutic antibodies targeting CD25 in HNSCC patients.

Anti-CCR4 Monoclonal Antibodies

The preferential expression of CCR4 on the surface of activated Tregs makes it a promising target [99]. Ex vivo depletion of CCR4+ T-cells from PBMCs of melanoma patients resulted in significant induction of antigen-specific effector T-cells [99]. Treatment of adult T-cell leukemia-lymphoma patients with anti-CCR4 mAb revealed depletion of Tregs and induction of antigen-specific T-cell response [99]. A phase I study of the same anti-CCR4 mAb KW-0761 (Mogamulizumab) in lung and esophageal cancer patients demonstrated effective Treg depletion from PBMCs following treatment [117]. Four phase I/II clinical trials evaluating anti-CCR4 antibody alone (NCT02281409) or in combination with other immune-activating therapies (NCT02301130, NCT02705105, NCT02444793 are currently recruiting patients with advanced solid cancers including HNSCC.

Anti-CTLA4 Monoclonal Antibodies

The anti-CTLA4 mAb Iplimumab is FDA-approved for the treatment of metastatic melanoma alone or in combination with PD checkpoint inhibition [118]. This strategy was originally thought to mediate its therapeutic effects through blockade of the inhibitory CTLA4 signal on the surface of effector T-cells. However, recent studies have demonstrated that anti-CTLA4-mAb may primarily mediate its anti-tumor effects through Treg depletion [119]. CTLA4+ Tregs have also been implicated in resistance of HNSCC patients to the anti-EGFR mAb cetuximab, suggesting a possible role for Iplimumab to improve responses to EGFR targeting and other therapies [120].

Anti-OX40 and GITR Agonist Antibodies

Tumor-derived Tregs commonly express two costimulatory molecules of the TNF receptor family; GITR and OX40 [121,122]. Although these receptors are considered positive costimulatory signals when found on the surface of effector T-cells and APCs, in the context of tumor-infiltrating Tregs they appear to play an inhibitory role leading to reduced Treg function and subsequent immune activation in the TME [122]. In HNSCC patients, tumor-infiltrating Tregs express a number of T-cell activation markers, including OX40 [123]. Ligation of OX40 on newly activated CD4 T-cells plays a role in their differentiation into effector T-cells or Tregs, depending upon the TME cytokine milieu [124]. Agonism of the OX40 receptor by either OX40L or an agonistic antibody improved survival and tumor-specific immunity in several mouse models – an effect which was initially only attributed to proliferation of effector and memory T-cells [122]. When anti-OX40 agonist antibody was studied in murine cancer models, the majority of tumors were rejected; this rejection was dependent upon OX40 agonism on both Tregs and effector T-cells [125]. In further preclinical studies, the combination of anti-OX40 agonist and the alkylating agent cyclophosphamide resulted in regression of established tumors and apoptosis of Tregs [126].

Two humanized anti-OX40 antibodies (MEDI6469, PF-04518600) are currently under study in phase I clinical trials. Studies of MEDI6469 in combination with radiation are underway in select tumor types (NCT01303705, NCT01862900, NCT02559024). A phase I trial of PF-04518600 monotherapy is currently recruiting patients with advanced/metastatic solid tumors (NCT02315066). In addition, a murine anti-OX40 antibody (MEDI6469) is currently in a phase I clinical trial as monotherapy prior to resection for locally advanced HNSCC (NCT02274155).

GITR is a co-stimulatory receptor found on the surface of APCs and CD4 T-cells [127]. Ligation of GITR with an agonist antibody directly inhibits naïve Treg-mediated immunosuppression in vitro, eradicates established tumors, and protects from melanoma cell line challenge in vivo [121,127,128]. However, it was initially unclear whether these anti-tumor effects were mediated by stimulation of effector T-cells, inhibition of Tregs, or both [128]. Recent work has confirmed that in vivo ligation of GITR leads to down-regulation of FoxP3 expression and reduced production of IL-10 by Tregs [129]. In addition, anti-GITR agonist antibody therapy synergizes with anti-CTLA4 blocking antibody in several solid tumor models [127,130]. Recent development of two humanized anti-GITR antibodies (TRX518, MEDI1873) has facilitated translation into phase I trials recruiting patients with advanced malignant melanoma or advanced solid tumors (NCT01239134, NCT02628574, NCT02583165).

Conclusions and Future Directions

With advances in our understanding of the intricacies of anti-tumor immunity, and the development and clinical application of checkpoint inhibitors and adoptive T-cell therapies, immunotherapy has taken center stage for many cancer types, including HNSCC. Advances in the clinical application of sequencing technology and MHC binding prediction algorithms will likely allow clinicians to identify immunotherapy-responsive HNSCC tumors in the treatment planning phase. Immunosuppression within the TME, mediated largely by tumor-infiltrating MDSCs, TAMs and Tregs, limits the efficacy of any type of immune activating therapy. While durable response rates of 15-30% with current immunotherapeutic strategies are significant, there is good evidence to support the concept that reversing local immunosuppression within the HNSCC TME could increase responses. Whether checkpoint inhibition or other immune activating approaches are utilized, future clinical trials will need to address the problem of local immunosuppression as a major limitation to the development of effective anti-tumor immunity. Challenges lie in how to best shift the balance of inflammation within the HNSCC TME toward effector immune cell activation and tumor rejection without damaging the anti-tumor immune response, itself, or inducing irreversible immune-related adverse events.

Highlights.

  1. The majority of head and neck cancers display high effector immune cell infiltration

  2. Head and neck cancer cells utilize many defined mechanisms to escape immune elimination

  3. An immunosuppressive immune infiltrate contributes heavily to local immune suppression

  4. Modulation of immune escape mechanisms may enhance responses to standard and immune therapies

Acknowledgements

We thank Anthony Saleh, PhD and Nicole Schmitt, MD for their critical appraisal of this manuscript.

Funding: This work was supported by National Institutes on Deafness and Other Communication Disorders intramural project number DC DC000087-01

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest: None

References

  • [1].Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517:576–82. doi: 10.1038/nature14129. doi:10.1038/nature14129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Ott PA, Adams S. Small-molecule protein kinase inhibitors and their effects on the immune system: implications for cancer treatment. Immunotherapy. 2011;3:213–27. doi: 10.2217/imt.10.99. doi:10.2217/imt.10.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Shimizu T, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, Smith LS, et al. The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer. Clin Cancer Res. 2012;18:2316–25. doi: 10.1158/1078-0432.CCR-11-2381. doi:10.1158/1078-0432.CCR-11-2381. [DOI] [PubMed] [Google Scholar]
  • [4].Sok JC, Coppelli FM, Thomas SM, Lango MN, Xi S, Hunt JL, et al. Mutant epidermal growth factor receptor (EGFRvIII) contributes to head and neck cancer growth and resistance to EGFR targeting. Clin Cancer Res. 2006;12:5064–73. doi: 10.1158/1078-0432.CCR-06-0913. doi:10.1158/1078-0432.CCR-06-0913. [DOI] [PubMed] [Google Scholar]
  • [5].Champiat S, Ferte C, Lebel-Binay S, Eggermont A, Soria JC. Exomics and immunogenics: Bridging mutational load and immune checkpoints efficacy. Oncoimmunology. 2014;3:e27817. doi: 10.4161/onci.27817. doi:10.4161/onci.27817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Keck MK, Zuo Z, Khattri A, Stricker TP, Brown CD, Imanguli M, et al. Integrative analysis of head and neck cancer identifies two biologically distinct HPV and three non-HPV subtypes. Clin Cancer Res. 2015;21:870–81. doi: 10.1158/1078-0432.CCR-14-2481. doi:10.1158/1078-0432.CCR-14-2481. [DOI] [PubMed] [Google Scholar]
  • [7].Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–8. doi: 10.1038/nature12213. doi:10.1038/nature12213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Wolf GT, Chepeha DB, Bellile E, Nguyen A, Thomas D, McHugh J. Tumor infiltrating lymphocytes (TIL) and prognosis in oral cavity squamous carcinoma: a preliminary study. Oral Oncol. 2015;51:90–5. doi: 10.1016/j.oraloncology.2014.09.006. doi:10.1016/j.oraloncology.2014.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Seiwert TY, Haddad RI, Gupta S, Mehra R, Tahara M, Berger R, et al. Antitumor activity and safety of pembrolizumab in patients (pts) with advanced squamous cell carcinoma of the head and neck (SCCHN): Preliminary results from KEYNOTE-012 expansion cohort. J Clin Oncol. 2015;33 doi: 10.1200/JCO.2016.68.1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Allen CT, Clavijo PE, Van Waes C, Chen Z. Anti-Tumor Immunity in Head and Neck Cancer: Understanding the Evidence, How Tumors Escape and Immunotherapeutic Approaches. Cancers (Basel) 2015;7:2397–414. doi: 10.3390/cancers7040900. doi:10.3390/cancers7040900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Mandruzzato S, Brasseur F, Andry G, Boon T, van der Bruggen P. A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma. J Exp Med. 1997;186:785–93. doi: 10.1084/jem.186.5.785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Yu P, Lee Y, Liu W, Chin RK, Wang J, Wang Y, et al. Priming of naive T cells inside tumors leads to eradication of established tumors. Nat Immunol. 2004;5:141–9. doi: 10.1038/ni1029. doi:10.1038/ni1029. [DOI] [PubMed] [Google Scholar]
  • [13].Smyth MJ, Crowe NY, Godfrey DI. NK cells and NKT cells collaborate in host protection from methylcholanthrene-induced fibrosarcoma. Int Immunol. 2001;13:459–63. doi: 10.1093/intimm/13.4.459. [DOI] [PubMed] [Google Scholar]
  • [14].Schantz SP, Shillitoe EJ, Brown B, Campbell B. Natural killer cell activity and head and neck cancer: a clinical assessment. J Natl Cancer Inst. 1986;77:869–75. [PubMed] [Google Scholar]
  • [15].Balermpas P, Michel Y, Wagenblast J, Seitz O, Weiss C, Rodel F, et al. Tumour-infiltrating lymphocytes predict response to definitive chemoradiotherapy in head and neck cancer. Br J Cancer. 2014;110:501–9. doi: 10.1038/bjc.2013.640. doi:10.1038/bjc.2013.640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Allen CT, Judd NP, Bui JD, Uppaluri R. The clinical implications of antitumor immunity in head and neck cancer. Laryngoscope. 2012;122:144–57. doi: 10.1002/lary.21913. doi:10.1002/lary.21913. [DOI] [PubMed] [Google Scholar]
  • [17].Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–92. doi: 10.1056/NEJMoa1113205. doi:10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].D’Amico L, Mahajan S, Capietto A-H, Yang Z, Zamani A, Ricci B, et al. Dickkopf-related protein 1 (Dkk1) regulates the accumulation and function of myeloid derived suppressor cells in cancer. J Exp Med. 2016 doi: 10.1084/jem.20150950. doi:10.1084/jem.20150950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Ariffin AB, Forde PF, Jahangeer S, Soden DM, Hinchion J. Releasing pressure in tumors: what do we know so far and where do we go from here? A review. Cancer Res. 2014;74:2655–62. doi: 10.1158/0008-5472.CAN-13-3696. doi:10.1158/0008-5472.CAN-13-3696. [DOI] [PubMed] [Google Scholar]
  • [20].Le QT, Shi G, Cao H, Nelson DW, Wang Y, Chen EY, et al. Galectin-1: a link between tumor hypoxia and tumor immune privilege. J Clin Oncol. 2005;23:8932–41. doi: 10.1200/JCO.2005.02.0206. doi:10.1200/JCO.2005.02.0206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].van Baren N, Van den Eynde BJ. Tumoral Immune Resistance Mediated by Enzymes That Degrade Tryptophan. Cancer Immunol Res. 2015:1–9. doi: 10.1158/2326-6066.CIR-15-0095. doi:10.1158/2326-6066.CIR-15-0095. [DOI] [PubMed] [Google Scholar]
  • [22].Lathers DM, Young MR. Increased aberrance of cytokine expression in plasma of patients with more advanced squamous cell carcinoma of the head and neck. Cytokine. 2004;25:220–8. doi: 10.1016/j.cyto.2003.11.005. doi:10.1016/j.cyto.2003.11.005. [DOI] [PubMed] [Google Scholar]
  • [23].Lu SL, Reh D, Li AG, Woods J, Corless CL, Kulesz-Martin M, et al. Overexpression of transforming growth factor beta1 in head and neck epithelia results in inflammation, angiogenesis, and epithelial hyperproliferation. Cancer Res. 2004;64:4405–10. doi: 10.1158/0008-5472.CAN-04-1032. doi:10.1158/0008-5472.CAN-04-1032. [DOI] [PubMed] [Google Scholar]
  • [24].Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell. 2015;162:1229–41. doi: 10.1016/j.cell.2015.08.016. doi:10.1016/j.cell.2015.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Ho PC, Bihuniak JD, Macintyre AN, Staron M, Liu X, Amezquita R, et al. Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses. Cell. 2015;162:1217–28. doi: 10.1016/j.cell.2015.08.012. doi:10.1016/j.cell.2015.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med. 2003;9:1269–74. doi: 10.1038/nm934. doi:10.1038/nm934. [DOI] [PubMed] [Google Scholar]
  • [27].Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nat Rev Immunol. 2008;8:467–77. doi: 10.1038/nri2326. doi:10.1038/nri2326. [DOI] [PubMed] [Google Scholar]
  • [28].Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol. 2012;24:207–12. doi: 10.1016/j.coi.2011.12.009. doi:10.1016/j.coi.2011.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Chen J, Feng Y, Lu L, Wang H, Dai L, Li Y, et al. Interferon-gamma-induced PD-L1 surface expression on human oral squamous carcinoma via PKD2 signal pathway. Immunobiology. 2012;217:385–93. doi: 10.1016/j.imbio.2011.10.016. doi:10.1016/j.imbio.2011.10.016. [DOI] [PubMed] [Google Scholar]
  • [30].Thomas GR, Chen Z, Leukinova E, Van Waes C, Wen J. Cytokines IL-1 alpha, IL-6, and GM-CSF constitutively secreted by oral squamous carcinoma induce down-regulation of CD80 costimulatory molecule expression: restoration by interferon gamma. Cancer Immunol Immunother. 2004;53:33–40. doi: 10.1007/s00262-003-0433-4. doi:10.1007/s00262-003-0433-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol. 2013;14:1014–22. doi: 10.1038/ni.2703. doi:10.1038/ni.2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–74. doi: 10.1038/nri2506. doi:10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Pak AS, Wright MA, Matthews JP, Collins SL, Petruzzelli GJ, Young MR. Mechanisms of immune suppression in patients with head and neck cancer: presence of CD34(+) cells which suppress immune functions within cancers that secrete granulocyte-macrophage colony-stimulating factor. Clin Cancer Res. 1995;1:95–103. [PubMed] [Google Scholar]
  • [34].Loukinova E, Dong G, Enamorado-Ayalya I, Thomas GR, Chen Z, Schreiber H, et al. Growth regulated oncogene-alpha expression by murine squamous cell carcinoma promotes tumor growth, metastasis, leukocyte infiltration and angiogenesis by a host CXC receptor-2 dependent mechanism. Oncogene. 2000;19:3477–86. doi: 10.1038/sj.onc.1203687. doi:10.1038/sj.onc.1203687. [DOI] [PubMed] [Google Scholar]
  • [35].Smith CW, Chen Z, Dong G, Loukinova E, Pegram MY, Nicholas-Figueroa L, et al. The host environment promotes the development of primary and metastatic squamous cell carcinomas that constitutively express proinflammatory cytokines IL-1alpha, IL-6, GM-CSF, and KC. Clin Exp Metastasis. 1998;16:655–64. doi: 10.1023/a:1006559811429. [DOI] [PubMed] [Google Scholar]
  • [36].Almand B, Clark JI, Nikitina E, van Beynen J, English NR, Knight SC, et al. Increased production of immature myeloid cells in cancer patients: a mechanism of immunosuppression in cancer. J Immunol. 2001;166:678–89. doi: 10.4049/jimmunol.166.1.678. [DOI] [PubMed] [Google Scholar]
  • [37].Weed DT, Vella JL, Reis IM, De la Fuente AC, Gomez C, Sargi Z, et al. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. 2015;21:39–48. doi: 10.1158/1078-0432.CCR-14-1711. doi:10.1158/1078-0432.CCR-14-1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Highfill SL, Cui Y, Giles AJ, Smith JP, Zhang H, Morse E, et al. Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci Transl Med. 2014;6:237ra67. doi: 10.1126/scitranslmed.3007974. doi:10.1126/scitranslmed.3007974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W, et al. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med. 2006;203:2691–702. doi: 10.1084/jem.20061104. doi:10.1084/jem.20061104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Katoh H, Wang D, Daikoku T, Sun H, Dey SK, Dubois RN. CXCR2-expressing myeloid-derived suppressor cells are essential to promote colitis-associated tumorigenesis. Cancer Cell. 2013;24:631–44. doi: 10.1016/j.ccr.2013.10.009. doi:10.1016/j.ccr.2013.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Lesokhin AM, Hohl TM, Kitano S, Cortez C, Hirschhorn-Cymerman D, Avogadri F, et al. Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment. Cancer Res. 2012;72:876–86. doi: 10.1158/0008-5472.CAN-11-1792. doi:10.1158/0008-5472.CAN-11-1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, et al. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74:5057–69. doi: 10.1158/0008-5472.CAN-13-3723. doi:10.1158/0008-5472.CAN-13-3723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Chen Z, Malhotra PS, Thomas GR, Ondrey FG, Duffey DC, Smith CW, et al. Expression of proinflammatory and proangiogenic cytokines in patients with head and neck cancer. Clin Cancer Res. 1999;5:1369–79. [PubMed] [Google Scholar]
  • [44].Keane MP, Belperio JA, Xue YY, Burdick MD, Strieter RM. Depletion of CXCR2 inhibits tumor growth and angiogenesis in a murine model of lung cancer. J Immunol. 2004;172:2853–60. doi: 10.4049/jimmunol.172.5.2853. [DOI] [PubMed] [Google Scholar]
  • [45].Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells-An overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology. 2015;4:e954829. doi: 10.4161/21624011.2014.954829. doi:10.4161/21624011.2014.954829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Lechner MG, Liebertz DJ, Epstein AL. Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J Immunol. 2010;185:2273–84. doi: 10.4049/jimmunol.1000901. doi:10.4049/jimmunol.1000901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Dong G, Chen Z, Kato T, Van Waes C. The host environment promotes the constitutive activation of nuclear factor-kappaB and proinflammatory cytokine expression during metastatic tumor progression of murine squamous cell carcinoma. Cancer Res. 1999;59:3495–504. [PubMed] [Google Scholar]
  • [48].Rodriguez PC, Hernandez CP, Quiceno D, Dubinett SM, Zabaleta J, Ochoa JB, et al. Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med. 2005;202:931–9. doi: 10.1084/jem.20050715. doi:10.1084/jem.20050715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Yu J, Du W, Yan F, Wang Y, Li H, Cao S, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190:3783–97. doi: 10.4049/jimmunol.1201449. doi:10.4049/jimmunol.1201449. [DOI] [PubMed] [Google Scholar]
  • [50].Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010;70:68–77. doi: 10.1158/0008-5472.CAN-09-2587. doi:10.1158/0008-5472.CAN-09-2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Vasquez-Dunddel D, Pan F, Zeng Q, Gorbounov M, Albesiano E, Fu J, et al. STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J Clin Invest. 2013;123:1580–9. doi: 10.1172/JCI60083. doi:10.1172/JCI60083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211:781–90. doi: 10.1084/jem.20131916. doi:10.1084/jem.20131916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Chikamatsu K, Sakakura K, Toyoda M, Takahashi K, Yamamoto T, Masuyama K. Immunosuppressive activity of CD14+ HLA-DR-cells in squamous cell carcinoma of the head and neck. Cancer Sci. 2012;103:976–83. doi: 10.1111/j.1349-7006.2012.02248.x. doi:10.1111/j.1349-7006.2012.02248.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Nagaraj S, Schrum AG, Cho H-I, Celis E, Gabrilovich DI. Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J Immunol. 2010;184:3106–16. doi: 10.4049/jimmunol.0902661. doi:10.4049/jimmunol.0902661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Nishikawa H, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Curr Opin Immunol. 2014;27:1–7. doi: 10.1016/j.coi.2013.12.005. doi:10.1016/j.coi.2013.12.005. [DOI] [PubMed] [Google Scholar]
  • [56].Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, et al. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66:1123–31. doi: 10.1158/0008-5472.CAN-05-1299. doi:10.1158/0008-5472.CAN-05-1299. [DOI] [PubMed] [Google Scholar]
  • [57].Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Kruger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology. 2008;135:234–43. doi: 10.1053/j.gastro.2008.03.020. doi:10.1053/j.gastro.2008.03.020. [DOI] [PubMed] [Google Scholar]
  • [58].Serafini P, Mgebroff S, Noonan K, Borrello I. Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer Res. 2008;68:5439–49. doi: 10.1158/0008-5472.CAN-07-6621. doi:10.1158/0008-5472.CAN-07-6621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Pan PY, Ma G, Weber KJ, Ozao-Choy J, Wang G, Yin B, et al. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010;70:99–108. doi: 10.1158/0008-5472.CAN-09-1882. doi:10.1158/0008-5472.CAN-09-1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Garrity T, Pandit R, Wright MA, Benefield J, Keni S, Young MR. Increased presence of CD34+ cells in the peripheral blood of head and neck cancer patients and their differentiation into dendritic cells. Int J Cancer. 1997;73:663–9. doi: 10.1002/(sici)1097-0215(19971127)73:5<663::aid-ijc9>3.0.co;2-v. [DOI] [PubMed] [Google Scholar]
  • [61].Lathers DM, Clark JI, Achille NJ, Young MR. Phase 1B study to improve immune responses in head and neck cancer patients using escalating doses of 25-hydroxyvitamin D3. Cancer Immunol Immunother. 2004;53:422–30. doi: 10.1007/s00262-003-0459-7. doi:10.1007/s00262-003-0459-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Walsh JE, Clark AM, Day TA, Gillespie MB, Young MR. Use of alpha,25-dihydroxyvitamin D3 treatment to stimulate immune infiltration into head and neck squamous cell carcinoma. Hum Immunol. 2010;71:659–65. doi: 10.1016/j.humimm.2010.04.008. doi:10.1016/j.humimm.2010.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Serafini P, Borrello I, Bronte V. Myeloid suppressor cells in cancer: recruitment, phenotype, properties, and mechanisms of immune suppression. Semin Cancer Biol. 2006;16:53–65. doi: 10.1016/j.semcancer.2005.07.005. doi:10.1016/j.semcancer.2005.07.005. [DOI] [PubMed] [Google Scholar]
  • [64].Okusaka T, Ueno H, Ikeda M, Mitsunaga S, Ozaka M, Ishii H, et al. Phase 1 and pharmacological trial of OPB-31121, a signal transducer and activator of transcription-3 inhibitor, in patients with advanced hepatocellular carcinoma. Hepatol Res. 2015 doi: 10.1111/hepr.12504. doi:10.1111/hepr.12504. [DOI] [PubMed] [Google Scholar]
  • [65].Hong DS, Younes A, Fayad L, Fowler NH, Hagemeister FB, Mistry R, et al. A phase I study of ISIS 481464 (AZD9150), a first-in-human, first-in-class, antisense oligonucleotide inhibitor of STAT3, in patients with advanced cancers. J Clin Oncol. 2013;31(suppl) abstr 8523. [Google Scholar]
  • [66].Seung LP, Rowley DA, Dubey P, Schreiber H. Synergy between T-cell immunity and inhibition of paracrine stimulation causes tumor rejection. Proc Natl Acad Sci U S A. 1995;92:6254–8. doi: 10.1073/pnas.92.14.6254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Srivastava MK, Zhu L, Harris-White M, Kar UK, Huang M, Johnson MF, et al. Myeloid suppressor cell depletion augments antitumor activity in lung cancer. PLoS One. 2012;7:e40677. doi: 10.1371/journal.pone.0040677. doi:10.1371/journal.pone.0040677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Stadtmann A, Zarbock A. CXCR2: From Bench to Bedside. Front Immunol. 2012;3:263. doi: 10.3389/fimmu.2012.00263. doi:10.3389/fimmu.2012.00263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Califano JA, Khan Z, Noonan KA, Rudraraju L, Zhang Z, Wang H, et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res. 2015;21:30–8. doi: 10.1158/1078-0432.CCR-14-1716. doi:10.1158/1078-0432.CCR-14-1716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].O’Sullivan T, Saddawi-Konefka R, Vermi W, Koebel CM, Arthur C, White JM, et al. Cancer immunoediting by the innate immune system in the absence of adaptive immunity. J Exp Med. 2012;209:1869–82. doi: 10.1084/jem.20112738. doi:10.1084/jem.20112738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Movahedi K, Laoui D, Gysemans C, Baeten M, Stangé G, Den Van Bossche J, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70:5728–39. doi: 10.1158/0008-5472.CAN-09-4672. doi:10.1158/0008-5472.CAN-09-4672. [DOI] [PubMed] [Google Scholar]
  • [72].Costa NL, Valadares MC, Souza PP, Mendonca EF, Oliveira JC, Silva TA, et al. Tumor-associated macrophages and the profile of inflammatory cytokines in oral squamous cell carcinoma. Oral Oncol. 2013;49:216–23. doi: 10.1016/j.oraloncology.2012.09.012. doi:10.1016/j.oraloncology.2012.09.012. [DOI] [PubMed] [Google Scholar]
  • [73].Marcus B, Arenberg D, Lee J, Kleer C, Chepeha DB, Schmalbach CE, et al. Prognostic factors in oral cavity and oropharyngeal squamous cell carcinoma. Cancer. 2004;101:2779–87. doi: 10.1002/cncr.20701. doi:10.1002/cncr.20701. [DOI] [PubMed] [Google Scholar]
  • [74].Galdiero MR, Bonavita E, Barajon I, Garlanda C, Mantovani A, Jaillon S. Tumor associated macrophages and neutrophils in cancer. Immunobiology. 2013;218:1402–10. doi: 10.1016/j.imbio.2013.06.003. doi:10.1016/j.imbio.2013.06.003. [DOI] [PubMed] [Google Scholar]
  • [75].Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers (Basel) 2014;6:1670–90. doi: 10.3390/cancers6031670. doi:10.3390/cancers6031670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Shah S, Cash H, Van Waes C, Allen C. Pools of programmed death ligand within the oral cavity tumor microenvironment are variable altered by targeted therapies. Head Neck. 2015 doi: 10.1002/hed.24269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61. doi: 10.1016/j.immuni.2014.06.010. doi:10.1016/j.immuni.2014.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Marchesi M, Andersson E, Villabona L, Seliger B, Lundqvist A, Kiessling R, et al. HLA-dependent tumour development: a role for tumour associate macrophages? J Transl Med. 2013;11:247. doi: 10.1186/1479-5876-11-247. doi:10.1186/1479-5876-11-247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Murai M, Turovskaya O, Kim G, Madan R, Karp CL, Cheroutre H, et al. Interleukin 10 acts on regulatory T cells to maintain expression of the transcription factor Foxp3 and suppressive function in mice with colitis. Nat Immunol. 2009;10:1178–84. doi: 10.1038/ni.1791. doi:10.1038/ni.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Chang LY, Lin YC, Mahalingam J, Huang CT, Chen TW, Kang CW, et al. Tumor-derived chemokine CCL5 enhances TGF-??-mediated killing of CD8 + T cells in colon cancer by T-regulatory cells. Cancer Res. 2012;72:1092–102. doi: 10.1158/0008-5472.CAN-11-2493. doi:10.1158/0008-5472.CAN-11-2493. [DOI] [PubMed] [Google Scholar]
  • [81].Chen KJ, Lin SZ, Zhou L, Xie HY, Zhou WH, Taki-Eldin A, et al. Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS One. 2011;6 doi: 10.1371/journal.pone.0024671. doi:10.1371/journal.pone.0024671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–9. doi: 10.1038/nm1093. doi:10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • [83].Mok S, Koya RC, Tsui C, Xu J, Robert L, Wu L, et al. Inhibition of CSF-1 receptor improves the antitumor efficacy of adoptive cell transfer immunotherapy. Cancer Res. 2014;74:153–61. doi: 10.1158/0008-5472.CAN-13-1816. doi:10.1158/0008-5472.CAN-13-1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011;1:54–67. doi: 10.1158/2159-8274.CD-10-0028. doi:10.1158/2159-8274.CD-10-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Ries CH, Cannarile MA, Hoves S, Benz J, Wartha K, Runza V, et al. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell. 2014;25:846–59. doi: 10.1016/j.ccr.2014.05.016. doi:10.1016/j.ccr.2014.05.016. [DOI] [PubMed] [Google Scholar]
  • [86].Dineen SP, Lynn KD, Holloway SE, Miller AF, Sullivan JP, Shames DS, et al. Vascular endothelial growth factor receptor 2 mediates macrophage infiltration into orthotopic pancreatic tumors in mice. Cancer Res. 2008;68:4340–6. doi: 10.1158/0008-5472.CAN-07-6705. doi:10.1158/0008-5472.CAN-07-6705. [DOI] [PubMed] [Google Scholar]
  • [87].Shime H, Matsumoto M, Oshiumi H, Tanaka S, Nakane a., Iwakura Y, et al. Toll-like receptor 3 signaling converts tumor-supporting myeloid cells to tumoricidal effectors. Proc Natl Acad Sci. 2012;109:2066–71. doi: 10.1073/pnas.1113099109. doi:10.1073/pnas.1113099109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Zhang X, Tian W, Cai X, Wang X, Dang W, Tang H, et al. Hydrazinocurcumin Encapsuled Nanoparticles “Re-Educate” Tumor-Associated Macrophages and Exhibit Anti-Tumor Effects on Breast Cancer Following STAT3 Suppression. PLoS One. 2013;8 doi: 10.1371/journal.pone.0065896. doi:10.1371/journal.pone.0065896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Coscia M, Quaglino E, Iezzi M, Curcio C, Pantaleoni F, Riganti C, et al. Zoledronic acid repolarizes tumour-associated macrophages and inhibits mammary carcinogenesis by targeting the mevalonate pathway. J Cell Mol Med. 2010;14:2803–15. doi: 10.1111/j.1582-4934.2009.00926.x. doi:10.1111/j.1582-4934.2009.00926.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Rolny C, Mazzone M, Tugues S, Laoui D, Johansson I, Coulon C, et al. HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PlGF. Cancer Cell. 2011;19:31–44. doi: 10.1016/j.ccr.2010.11.009. doi:10.1016/j.ccr.2010.11.009. [DOI] [PubMed] [Google Scholar]
  • [91].Downey CM, Aghaei M, Schwendener RA, Jirik FR. DMXAA causes tumor site-specific vascular disruption in murine non-small cell lung cancer, and like the endogenous non-canonical cyclic dinucleotide STING agonist, 2′3′-cGAMP, induces M2 macrophage repolarization. PLoS One. 2014;9 doi: 10.1371/journal.pone.0099988. doi:10.1371/journal.pone.0099988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Baird JR, Friedman D, Cottam B, Dubensky TW, Kanne DB, Bambina S, et al. Radiotherapy Combined with Novel STING-Targeting Oligonucleotides Results in Regression of Established Tumors. Cancer Res. 2015;76:50–61. doi: 10.1158/0008-5472.CAN-14-3619. doi:10.1158/0008-5472.CAN-14-3619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Weed DT, Walker G, De La Fuente AC, Nazarian R, Vella JL, Gomez-Fernandez CR, et al. FOXP3 subcellular localization predicts recurrence in oral squamous cell carcinoma. PLoS One. 2013;8:e71908. doi: 10.1371/journal.pone.0071908. doi:10.1371/journal.pone.0071908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Badoual C, Hans S, Rodriguez J, Peyrard S, Klein C, Agueznay Nel H, et al. Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancers. Clin Cancer Res. 2006;12:465–72. doi: 10.1158/1078-0432.CCR-05-1886. doi:10.1158/1078-0432.CCR-05-1886. [DOI] [PubMed] [Google Scholar]
  • [95].Strauss L, Bergmann C, Gooding W, Johnson JT, Whiteside TL. The frequency and suppressor function of CD4+CD25highFoxp3+ T cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clin Cancer Res. 2007;13:6301–11. doi: 10.1158/1078-0432.CCR-07-1403. doi:10.1158/1078-0432.CCR-07-1403. [DOI] [PubMed] [Google Scholar]
  • [96].Zhou G, Levitsky HI. Natural Regulatory T Cells and De Novo-Induced Regulatory T Cells Contribute Independently to Tumor-Specific Tolerance. J Immunol. 2007;178:2155–62. doi: 10.4049/jimmunol.178.4.2155. doi:10.4049/jimmunol.178.4.2155. [DOI] [PubMed] [Google Scholar]
  • [97].Valzasina B, Piconese S, Guiducci C, Colombo MP. Tumor-induced expansion of regulatory T cells by conversion of CD4+CD25− lymphocytes is thymus and proliferation independent. Cancer Res. 2006;66:4488–95. doi: 10.1158/0008-5472.CAN-05-4217. doi:10.1158/0008-5472.CAN-05-4217. [DOI] [PubMed] [Google Scholar]
  • [98].Jie HB, Gildener-Leapman N, Li J, Srivastava RM, Gibson SP, Whiteside TL, et al. Intratumoral regulatory T cells upregulate immunosuppressive molecules in head and neck cancer patients. Br J Cancer. 2013;109:2629–35. doi: 10.1038/bjc.2013.645. doi:10.1038/bjc.2013.645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Sugiyama D, Nishikawa H, Maeda Y, Nishioka M, Tanemura A, Katayama I, et al. Anti-CCR4 mAb selectively depletes effector-type FoxP3+CD4+ regulatory T cells, evoking antitumor immune responses in humans. Proc Natl Acad Sci U S A. 2013;110:17945–50. doi: 10.1073/pnas.1316796110. doi:10.1073/pnas.1316796110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Facciabene A, Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. 2011;475:226–30. doi: 10.1038/nature10169. doi:10.1038/nature10169. [DOI] [PubMed] [Google Scholar]
  • [101].Redjimi N, Raffin C, Raimbaud I, Pignon P, Matsuzaki J, Odunsi K, et al. CXCR3+ T regulatory cells selectively accumulate in human ovarian carcinomas to limit type I immunity. Cancer Res. 2012;72:4351–60. doi: 10.1158/0008-5472.CAN-12-0579. doi:10.1158/0008-5472.CAN-12-0579. [DOI] [PubMed] [Google Scholar]
  • [102].Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007;204:1757–64. doi: 10.1084/jem.20070590. doi:10.1084/jem.20070590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–85. doi: 10.1084/jem.20070602. doi:10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Perez N, Karumuthil-Melethil S, Li R, Prabhakar BS, Holterman MJ, Vasu C. Preferential costimulation by CD80 results in IL-10-dependent TGF-beta1(+) -adaptive regulatory T cell generation. J Immunol. 2008;180:6566–76. doi: 10.4049/jimmunol.180.10.6566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Pallandre JR, Brillard E, Crehange G, Radlovic A, Remy-Martin JP, Saas P, et al. Role of STAT3 in CD4+CD25+FOXP3+ regulatory lymphocyte generation: implications in graft-versus-host disease and antitumor immunity. J Immunol. 2007;179:7593–604. doi: 10.4049/jimmunol.179.11.7593. [DOI] [PubMed] [Google Scholar]
  • [106].Ahmadzadeh M, Rosenberg SA. IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood. 2006;107:2409–14. doi: 10.1182/blood-2005-06-2399. doi:10.1182/blood-2005-06-2399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Bonertz A, Weitz J, Pietsch DH, Rahbari NN, Schlude C, Ge Y, et al. Antigen-specific Tregs control T cell responses against a limited repertoire of tumor antigens in patients with colorectal carcinoma. J Clin Invest. 2009;119:3311–21. doi: 10.1172/JCI39608. doi:10.1172/JCI39608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Nishikawa H, Kato T, Tawara I, Takemitsu T, Saito K, Wang L, et al. Accelerated chemically induced tumor development mediated by CD4+CD25+ regulatory T cells in wild-type hosts. Proc Natl Acad Sci U S A. 2005;102:9253–7. doi: 10.1073/pnas.0503852102. doi:10.1073/pnas.0503852102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Strauss L, Bergmann C, Szczepanski M, Gooding W, Johnson JT, Whiteside TL. A unique subset of CD4+CD25highFoxp3+ T cells secreting interleukin-10 and transforming growth factor-beta1 mediates suppression in the tumor microenvironment. Clin Cancer Res. 2007;13:4345–54. doi: 10.1158/1078-0432.CCR-07-0472. doi:10.1158/1078-0432.CCR-07-0472. [DOI] [PubMed] [Google Scholar]
  • [110].Bergmann C, Strauss L, Wang Y, Szczepanski MJ, Lang S, Johnson JT, et al. T regulatory type 1 cells in squamous cell carcinoma of the head and neck: mechanisms of suppression and expansion in advanced disease. Clin Cancer Res. 2008;14:3706–15. doi: 10.1158/1078-0432.CCR-07-5126. doi:10.1158/1078-0432.CCR-07-5126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Cao X, Cai SF, Fehniger TA, Song J, Collins LI, Piwnica-Worms DR, et al. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity. 2007;27:635–46. doi: 10.1016/j.immuni.2007.08.014. doi:10.1016/j.immuni.2007.08.014. [DOI] [PubMed] [Google Scholar]
  • [112].Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, et al. CTLA-4 control over Foxp3+ regulatory T cell function. Science (80−) 2008;322:271–5. doi: 10.1126/science.1160062. doi:10.1126/science.1160062. [DOI] [PubMed] [Google Scholar]
  • [113].Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204:1257–65. doi: 10.1084/jem.20062512. doi:10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Jacobs JF, Punt CJ, Lesterhuis WJ, Sutmuller RP, Brouwer HM, Scharenborg NM, et al. Dendritic cell vaccination in combination with anti-CD25 monoclonal antibody treatment: a phase I/II study in metastatic melanoma patients. Clin Cancer Res. 2010;16:5067–78. doi: 10.1158/1078-0432.CCR-10-1757. doi:10.1158/1078-0432.CCR-10-1757. [DOI] [PubMed] [Google Scholar]
  • [115].Rech AJ, Mick R, Martin S, Recio A, Aqui NA, Powell DJ, Jr., et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci Transl Med. 2012;4:134ra62. doi: 10.1126/scitranslmed.3003330. doi:10.1126/scitranslmed.3003330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Whelan MC, Casey G, Larkin JO, Guinn BA, O’Sullivan GC, Tangney M. Oral tolerance to cancer can be abrogated by T regulatory cell inhibition. PLoS One. 2014;9:e97602. doi: 10.1371/journal.pone.0097602. doi:10.1371/journal.pone.0097602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Kurose K, Ohue Y, Wada H, Iida S, Ishida T, Kojima T, et al. Phase Ia Study of FoxP3+ CD4 Treg Depletion by Infusion of a Humanized Anti-CCR4 Antibody, KW-0761, in Cancer Patients. Clin Cancer Res. 2015;21:4327–36. doi: 10.1158/1078-0432.CCR-15-0357. doi:10.1158/1078-0432.CCR-15-0357. [DOI] [PubMed] [Google Scholar]
  • [118].Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–23. doi: 10.1056/NEJMoa1003466. doi:10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Selby MJ, Engelhardt JJ, Quigley M, Henning KA, Chen T, Srinivasan M, et al. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol Res. 2013;1:32–42. doi: 10.1158/2326-6066.CIR-13-0013. doi:10.1158/2326-6066.CIR-13-0013. [DOI] [PubMed] [Google Scholar]
  • [120].Jie HB, Schuler PJ, Lee SC, Srivastava RM, Argiris A, Ferrone S, et al. CTLA-4(+) Regulatory T Cells Increased in Cetuximab-Treated Head and Neck Cancer Patients Suppress NK Cell Cytotoxicity and Correlate with Poor Prognosis. Cancer Res. 2015;75:2200–10. doi: 10.1158/0008-5472.CAN-14-2788. doi:10.1158/0008-5472.CAN-14-2788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Turk MJ, Guevara-Patino JA, Rizzuto GA, Engelhorn ME, Sakaguchi S, Houghton AN. Concomitant tumor immunity to a poorly immunogenic melanoma is prevented by regulatory T cells. J Exp Med. 2004;200:771–82. doi: 10.1084/jem.20041130. doi:10.1084/jem.20041130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Weinberg AD, Rivera MM, Prell R, Morris A, Ramstad T, Vetto JT, et al. Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol. 2000;164:2160–9. doi: 10.4049/jimmunol.164.4.2160. [DOI] [PubMed] [Google Scholar]
  • [123].Montler R, Bell RB, Thalhofer C, Leidner R, Feng Z, Fox BA, et al. OX40, PD-1 and CTLA-4 are selectively expressed on tumor-infiltrating T cells in head and neck cancer. Clin Transl Immunol. 2016;5:e70. doi: 10.1038/cti.2016.16. doi:10.1038/cti.2016.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Bell RB, Leidner RS, Crittenden MR, Curti BD, Feng Z, Montler R, et al. OX40 signaling in head and neck squamous cell carcinoma: Overcoming immunosuppression in the tumor microenvironment. Oral Oncol. 2016;52:1–10. doi: 10.1016/j.oraloncology.2015.11.009. doi:10.1016/j.oraloncology.2015.11.009. [DOI] [PubMed] [Google Scholar]
  • [125].Piconese S, Valzasina B, Colombo MP. OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med. 2008;205:825–39. doi: 10.1084/jem.20071341. doi:10.1084/jem.20071341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Hirschhorn-Cymerman D, Rizzuto GA, Merghoub T, Cohen AD, Avogadri F, Lesokhin AM, et al. OX40 engagement and chemotherapy combination provides potent antitumor immunity with concomitant regulatory T cell apoptosis. J Exp Med. 2009;206:1103–16. doi: 10.1084/jem.20082205. doi:10.1084/jem.20082205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, Yamaguchi T, et al. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J Exp Med. 2005;202:885–91. doi: 10.1084/jem.20050940. doi:10.1084/jem.20050940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Shimizu J, Yamazaki S, Takahashi T, Ishida Y, Sakaguchi S. Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol. 2002;3:135–42. doi: 10.1038/ni759. doi:10.1038/ni759. [DOI] [PubMed] [Google Scholar]
  • [129].Schaer DA, Budhu S, Liu C, Bryson C, Malandro N, Cohen A, et al. GITR pathway activation abrogates tumor immune suppression through loss of regulatory T cell lineage stability. Cancer Immunol Res. 2013;1:320–31. doi: 10.1158/2326-6066.CIR-13-0086. doi:10.1158/2326-6066.CIR-13-0086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Mitsui J, Nishikawa H, Muraoka D, Wang L, Noguchi T, Sato E, et al. Two distinct mechanisms of augmented antitumor activity by modulation of immunostimulatory/inhibitory signals. Clin Cancer Res. 2010;16:2781–91. doi: 10.1158/1078-0432.CCR-09-3243. doi:10.1158/1078-0432.CCR-09-3243. [DOI] [PubMed] [Google Scholar]

RESOURCES