Cancer Metastasis Rev DOI 10.1007/s10555-015-9566-0

Regulatory T cells and potential inmmunotherapeutic targets in lung cancer Ding Zhang 1 & Zhihong Chen 1 & Diane C. Wang 1 & Xiangdong Wang 1

# Springer Science+Business Media New York 2015

Abstract Lung cancer and metastasis are two of the most lethal diseases globally and seldom have effective therapies. Immunotherapy is considered as one of the powerful alternatives. Regulatory T cells (Tregs) can suppress the activation of the immune system, maintain immune tolerance to self-antigens, and contribute to immunosuppression of antitumor immunity, which is critical for tumor immune evasion in epithelial malignancies, including lung cancer. The present review gives an overview of the biological functions and regulations of Tregs associated with the development of lung cancer and metastasis and explores the potentials of Treg-oriented therapeutic targets. Subsets and features of Tregs mainly include naturally occurring Tregs (nTregs) (CD4+ nTregs and CD8+ nTregs) and adaptive/induced Tregs (CD4+ iTregs and CD8+ iTregs). Tregs, especially in circulation or regional lymph nodes, play an important role in the progress and metastasis of lung cancer and are considered as therapeutic targets and biomarkers to predict the survival length and recurrence of lung cancer. Increasing understanding of Tregs’ functional mechanisms will lead to a number of clinical trials on the discovery and development of Treg-oriented new therapies. Tregs play important roles in lung cancer and metastasis, and the understanding of Tregs becomes more critical for clinical applications and therapies. Thus, Tregs and associated

Ding Zhang and Zhihong Chen contributed equally to this work. * Xiangdong Wang [email protected] 1

Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Shanghai, China

factors can be potential therapeutic targets for lung cancer immunotherapy. Keywords Lung cancer . Regulatory Tcell . Immunotherapy . Metastasis

1 Introduction Lung cancer and metastasis are responsible for the most lethality of diseases [1, 2],with about 15 % overall 5-year survival rates for patients with lung cancer. The immune system protects the host against tumor development, growth, and metastasis [3–5]. Regulatory T cells (Tregs) are a population of T cells suppressing the activation of the immune system and maintaining immune tolerance to self-antigens. Tregs can contribute to immunosuppression of antitumor immunity, allowing the tumor evasion through the immune barrier in epithelial malignancies [6]. Tregs were upregulated or activated in the tumor microenvironment, where increased number of Tregs was correlated with a poor prognosis in epithelial cancers, including lung cancer [7, 8]. The present review gives an overview of the biological functions and regulations of Tregs associated with the development of lung cancer and metastasis and explores the potentials of Treg-oriented therapeutic targets. The present review aims at defining subsets and features of Tregs, potential roles of Tregs from circulation or regional lymph nodes in progress and metastasis of lung cancer, and therapeutic targets or biomarkers to predict the survival length and recurrence of lung cancer. Tregs may suppress the function of immune cells and allow lung cancer escapes probably through cell interaction-dependent or cytokine-mediated suppression, although the exact mechanism of Treg cell-mediated suppression remains unclear.

Cancer Metastasis Rev

2 Subsets and features of Tregs Tregs were initially described as thymus-derived suppressive T cells in the early 1970s [9], to probably interconnect between Bsuppressor cells^ and malignancies [10]. Several phenotypical and functional subsets of distinct Tregs with CD4 and CD8 were described, after CD4+CD25+ T cells were then named as thymus-derived naturally occurring regulatory T cells (nTregs) since 1995 [11]. Tregs are recently divided into naturally occurring Tregs (nTregs) (CD4+ nTregs and CD8+ nTregs) and adaptive/induced Tregs (CD4+ iTregs and CD8+ iTregs) (Fig. 1). 2.1 CD4+ CD25+ nTregs CD4+ CD25+ nTregs are derived from the thymus and represent about 1–2 % of the total peripheral CD4+ T-cell population in human [12], in the control of immune self-tolerance, allograft rejection and allergy, or inhibition of the effector functions during infection and in tumors. CD25 is the αchain of the high-affinity receptor for interleukin-2. The activation of nTregs depends upon IL-2 production and concentration within the microenvironment [13]. Forkhead Box protein 3 (FOXP3) was identified as an important regulator of nTreg development and function [11], while Tregs constitutively express cytotoxic T lymphocyte antigen-4 (CTLA-4) [14] and glucocorticoid-induced tumor necrosis factor receptor family-related gene (GITR) [15]. Few biological functionspecific biomarkers were still discovered to present functional characters of those cells [16]. To discriminate Tregs from conventional (activated) CD4+ T cells, a low expression of CD127 (the α-chain of the IL-7 receptor) and a modulated CD45RB expression were proposed as comarkers together with the expression of FOXP3 and CD25 [17, 18]. Helios, a member of the Ikaros transcription factor family, was recently Fig. 1 Subsets and suppressive mechanisms of Tregs. Tregs are mainly divided into nTregs and iTregs with CD4+ or CD8+ subsets. Both can play the immunosuppressive roles in cell interaction-dependent and/or mediator-dependent manners (by secreting IL-10, IL-35, TGF-β, or other soluble factors). nTregs naturally occurring regulatory T cells, iTregs induced regulatory T cells, Tr1 IL-10-secreting regulatory T cell, Th3 TGF-βsecreting regulatory T cell, Tr35 IL-35-secreting regulatory T cell, IL interleukin, TGF-β transforming growth factor-β

proposed as a potentially specific marker of thymus-derived nTregs [19], although it is still questioned by other studies [20]. Circulating human antigen-reactive CD4+FOXP3+ nTregs or CD4+CD25+FOXP3+ nTregs were identified, isolated, and expanded with minimal loss of functional activity [21]. 2.2 Induced (adaptive) CD4+ regulatory T cells iTregs derived from naive CD4+ T cells in the periphery modulate the immune response to microbial and tissue antigens, different from nTregs which developed in the thymus that play a critical role in regulating self-tolerance and seed peripheral tissues to suppress the activation of effector cells [22, 23]. Subtypes of CD4+ iTregs include IL-10-secreting T regulatory 1 (Tr1) cells, TGF-β-secreting T regulatory (Th3) cells, or CD25+FOXP3+ nTreg-like cells. Tr1 cells produce a high amount of IL-10 and little amounts of FOXP3 and CD25 under the stimulation with immature dendritic cells (DCs) in the presence of IL-10 [24]. Foxp3+ and Foxp3− precursor cells give rise to peripheral Tr1 by a mechanism dependent on TGF-β rather than IL-10 [25]. It indicated that human Tr1 may be an important and clinically feasible target in cancer patients for therapy [26], even though Tr1 Tregs were believed more functionally specialized in the regulation of inflammation [27]. Th3 cells, initially described in the induction of oral tolerance to food antigens [28], may mediate Bbystander suppression^ when encountering the fed autoantigen at the target organ [29]. TGF-β can induce the differentiation of Th3 cells and suppress T-cell responses through the induction of Tregs, rather than direct inhibition of cell proliferation [30]. CD4+CD25+FOXP3+ iTreg-like cells are generated in peripheral lymphoid tissues by the conversion of conventional naive CD4+FoxP3− T cells to FoxP3+ Tregs with polyclonal or antigen-specific activation in the presence of specific immune

Cancer Metastasis Rev

suppressive cytokines [31]. The minimal program for Foxp3+ iTreg development and differentiation requires the stimulation and the appearance of cytokines TGF-β and IL-2 that generated iTregs in mesenteric lymph nodes, during induction of oral tolerance, chronically inflamed tissues, or tumor [31–33]. 2.3 CD8+ nTregs or CD8+ iTregs CD8+ Tregs are divided into naturally occurring and induced CD8+ regulatory T cells, i.e., CD8+ nTregs and CD8+ iTregs, respectively, to mildly suppress T-cell proliferation and interferon-γ production [34], although CD8+ nTregs and CD8+ iTregs express CD25, GITR, CTLA4, and CD103. Several phenotypes of CD8+ iTregs have been identified in peripheral tissue in varied conditions. CD8+ iTregs were also discovered in cancer patients, e.g., ovarian, prostate, or nasopharyngeal carcinoma, and the roles of CD8+ iTregs in cancer are emphasized [35].

3 Tregs in progress and metastasis of lung cancer Several studies demonstrated that tumor-associated T cells from patients with epithelial tumors at early and late stages contained increased proportions of CD4+CD25+ Tregs in the peripheral blood. Those cells secreted immunosuppressive cytokines, e.g., TGF-β, and expressed high levels of CTLA-4 on the cell surface to contribute to Treg-associated immune dysfunction in patients with nonsmall cell lung cancer (NSCLC) [36–40]. CD4+CD25+ Tregs in tumor-infiltrating lymph nodes from patients with NSCLC increased significantly, as compared with tumor-free lymph nodes. The percentage of CD4+CD25+ Tregs in tumor-infiltrating lymph nodes of NSCLC patients was negatively correlated with the amount with CD8+ cells within lymph nodes while positively with the levels of TGF-β1 or IL-10 [41]. CD13+CD4+CD25hi Treg cells as an important subpopulation among CD4+CD25hi Treg cells were identified in the peripheral blood of NSCLC patients [42] and expressed higher levels of Foxp3, CTLA-4, membrane-bound transforming growth factor β1, and programmed death legend-1 (PD-L1). CD13+CD4+CD25hi Treg cells could suppress CD25 expression and proliferation of CD4+CD25− T cells. The percentage of CD13+CD4+CD25hi Tregs among CD4+CD25hi Tregs correlated with pathological stages of NSCLC and tumor burden. The percentage of CD8+CD28− Tregs was also higher in the peripheral blood of patients with lung cancer [39, 43]. Increased Tregs in the circulation were found to be associated with worse overall and relapse-free survivals, tumor FOXP3 expression with better prognosis in NSCLC, tumor-infiltrating Tregs, or high-risks patients [8]. In addition, the reciprocal CD4+ T-cell balance bet w e e n e ff e c t o r C D 6 2 L l o w C D 4 + T e ff e c t o r a n d

CD62LhighCD25+CD4+ Tregs may reflect the stage of small cell lung cancer (SCLC) and predict the survival length and recurrence [44]. Local and systemic immune responses in SCLC patients might be downregulated by the differentiation of functional CD4+CD25+FOXP3+CD127loHeliosTregs, partially depending on IL-15 [45]. Hasegawa et al. [46] analyzed peripheral blood mononuclear cells collected prior to surgery and resected specimens from patients with NSCLC and found that peripheral Tregs (pTregs) were CD4+ and Foxp3+. CD4, CD8, and Foxp3 expressions were observed in central regions as cCD4, cCD8, and cFoxp3 or interstitial regions of the tumor as iCD4, iCD8, and iFoxp3. The higher frequency of pTregs was noticed in patients with pleural, vessel, or lymphatic vessel invasions, or with recurrent cancer. Higher frequencies of cCD4 or cCD8 were detected in patients with T1 cancer. Cancer patients with low levels of cFoxp3/iFoxp3 had higher frequency of pTregs as an independent prognostic factor. It implies that pTregs play an important role in the development of lung cancer recurrence and metastasis. The proportion of Tregs in regional lymph node lymphocytes was higher than in peripheral lymphocytes in NSCLC patients as poor prognostic factor in patients with or without node metastasis, associated with lower 5-year overall survival rates [47]. It was also found that the proportion of Tregs in regional lymph node lymphocytes was higher in patients with lung adenocarcinoma, as compared with lung inflammation or squamous carcinomas [48]. It suggests that Treg target therapy may benefit patients with lung adenocarcinoma. The number of Tregs in tumor stroma was higher than in tumor nest in pathological stage I lung invasive adenocarcinoma. The higher Tregs the stroma had, the poorer the prognosis of patients was. Cancer-associated fibroblasts (CAF) could influence Treg induction probably through TGF-β, vascular endothelial growth factor, or both [49]. It is possible that CAFgenerated immune-regulatory cytokines may induce Tregs in the stroma, forming a tumor-promoting microenvironment in lung adenocarcinoma and leading to poor outcome. In addition, docetaxel-based chemotherapy could reduce nonsuppressive, resting, or activated Treg subsets after 4 cycles of treatment in NSCLC patients, probably through a direct effect [50]. Those lines of evidence suggest that Tregs are involved in the regulation of local antitumor immunity, promotion of cancer progression and metastasis, or patient prognosis.

4 Mechanisms of Treg-targeting immunotherapy for lung cancer Tregs may suppress the function of innate and adaptive immune cells through cell interaction-dependent suppression or cytokine-mediated suppression [51], although the exact mechanism of Treg cell-mediated suppression remains unclear. The

Cancer Metastasis Rev

present review mainly focused on mechanisms of Tregmediated immune suppression and potential Treg-targeting immunotherapy for lung cancer. 4.1 Cell interaction-dependent suppression Tregs can suppress directly immune cells by cell-cell contacts via cell surface molecules such as CTLA-4, programmed death 1 (PD-1)/PD-L1, lymphocyte activation gene (LAG3), GITR, CD39/CD73, CD95–CD95 ligand, granzymes/ perforin, or neuropilin-1 (Nrp-1), as shown in Fig. 2. CTLA4 is homologous to CD28, located on the surface of T cells and expressed highly on Tregs. CTLA-4 binds with higher affinity to B7-1 and B7-2 than CD28 and is more competitive [52]. Such signal of naive T-cell activation in response to tumor cell invasion can be influenced, and Treg-mediated suppression of effector T cells via DCs was reduced by blocking or deleting CTLA-4 [14, 53]. It suggests that targeting CTLA-4 can be an anticancer strategy. Recently, an anti-CTLA-4 agent (Yervoy, ipilimumab) was approved as a new immunotherapy for metastatic melanoma [54]. Tregs increased in metastatic stage and CTLA-4 was overexpressed in the lymphocytes of patients with NSCLC [55]. CTLA-4 and CD28 gene polymorphism was associated with NSCLC [56]. CTLA-4 could induce DCs to express indoleamine 2,3-dioxygenase via interaction between CD80 and CD86, to induce the catabolism of tryptophan into proapoptotic metabolites and suppress effector T cells [57]. It suggests that immunotherapy regimen targeting CTLA-4 of Tregs might benefit lung cancer patients, although

Fig. 2 Contact-dependent suppression and potential targets of Tregs for lung cancer immunotherapy. a CTLA-4 binds to B7.1/2 costimulatory molecules on DCs with CD28 on T cells; b LAG-3 binds MHC class II molecules expressed on DCs with high affinity; c agonist anti-GITR antibodies could overcome self-tolerance and reverse Treg suppression; d the CD39 ectoenzyme produces AMP from ATP or ADP, which is subsequently converted into extracellular adenosine by the CD73 ectoenzyme. Adenosine binding to ADORA2A on activated T effector cells or NKs generates immunosuppressive effect; e exogenous CD95 results in a systemic decrease of Tregs through AICD; f CD4+CD25+

the overexpression of CTLA-4 in radically resected NSCLC indicates a reduced death rate [58]. The blocking of CTLA-4 permitted antitumor T cells to acquire effector function and improved the survival rate, evidenced by randomized phase III studies in patients with metastatic cancer [59–61]. Long-term follow-up from a clinical trial showed that a 5-year survival rate was approximately 15 % of treated patients after enrollment [60]. CTLA-4 blockade is moving forward in lung cancer because the immune checkpoint molecule likely evolved to protect self-tissues from autoimmunity. A phase II study in NSCLC on the activity of ipilimumab plus paclitaxel or carboplatin showed that phased ipilimumab plus paclitaxel or carboplatin improved immune-related progression-free survival and progression-free survival [62]. Abscopal responses of NSCLC patients led to a posttreatment increase in tumorinfiltrating cytotoxic lymphocytes, tumor regression, or normalization of tumor markers 1 year after treatment with concurrent radiotherapy and ipilimumab [63]. Another phase II study in extensive disease SCLC showed that phased ipilimumab rather than concurrent ipilimumab improved immune-related progression-free survival while not progression-free survival [64], although results from some phase II trials of ipilimumab in advanced NSCLC and SCLC are encouraging and phase III trials are still ongoing. PD-1, one of the immune checkpoint molecules, is expressed on activated and exhausted peripheral T cells in lung cancer and on tumor cells, and PD-1/PD-L1 interaction provided a negative signal for antigen-induced T-cell activation [65–67]. PD-1 expressed on tumor-infiltrating T cells

Tregs and CD4 + CD25 − T cells regulate death/growth arrest by differentially utilizing the granzyme–perforin pathway. LL-37 regulates CD4+CD25+FoxP3+ Tregs through apoptosis; g Nrp-1 expresses on Tregs and promotes interactions with immature DCs. DCs dendritic cells, CTLA-4 cytotoxic T lymphocyte antigen-4, LAG-3 lymphocyte activation gene 3, GITR glucocorticoid-induced tumor necrosis factor receptor, ADORA2A adenosine A2A receptor, AICD activation-induced cell death, LL-37 a human cathelicidin, Nrp-1 neuropilin-1, LAP latent form of TGF-β

Cancer Metastasis Rev

were correlated with the prognosis of patients with NSCLC, which could be influenced by PD-1 blockade on a single agent [68–70]. Antibodies against PD-1 have recently entered phase III clinical trials, where the rate of adverse events at grades 3 and 4 was lower in PD-1 blockade than in CTLA-4 blockade [71]. The PD-1/PD-ligand pathway may act more peripherally than the CTLA-4/B7-1 pathway which may play a role in lymph nodes. Clinical studies on nivolumab against PD-1 and ipilimumab showed that the combination may provide a feasible regimen as the first-line treatment for chemotherapynaive patients with NSCLC [72]. LAG-3 (CD223), a CD4 homologue, can bind major histocompatibility complex (MHC) class II molecules with high affinity, for maximal suppressive activity of nTregs and iTregs [73]. The depletion of LAG-3+CD4+ T cells enhanced tumor-specific CD8 + T-cell reactivity, and LAG-3–Ig fusion protein mediated tumor control and regression [74, 75]. Based on the targeting potential of LAG-3–Ig, a new drug (IMP321, Immutep, Paris) was developed and tested in clinical trials. IMP321 is well tolerated and associated with an enhanced TH1 CD4+ T cell and CD8+ T cell [76, 77]. The combination of IMP321 with taxane-based chemotherapy in women with breast cancer demonstrated a promisingly objective response rate [78]. A recent study found an enhanced suppressive capacity of LAG-3+CD4+CD25+ T cells, as compared with LAG-3− T cells from tumor sites of patients [79]. It indicates that LAG-3 could be a promising target in lung cancer immunotherapy, although further studies are still needed to clarify the role of LAG-3 in lung cancer. GITR is considered as a target for cancer immunotherapy on the basis of the evidence that agonist anti-GITR antibodies could overcome self-tolerance and reverse Treg suppression [80]. Therapeutic effects of anti-GITR antibodies were primarily demonstrated in melanoma [81], colon tumors [82], lymphoma [83], or vaccine-treated-tumors [84]. The combination of DTA-1 with adoptive T-cell therapy or CTLA-4 blockade could synergistically enhance tumor regression of fibrosarcoma and colon tumors [85, 86]. The addition of DCs expressing anti-CTLA-4 to the original glucocorticoid-induced tumor necrosis factor receptor ligand (GITRL) and TYRP-2 expressing DC vaccine further enhanced the efficacy of the approach [87]. The agonist anti-GITR antibodies in tumor immunotherapy could make T effectors resistant to Treg suppression [88], indicating that GITR may have different effects on intratumor and peripheral Tregs [89]. GITR-induced changes in nTreg phenotype and function may depend upon phosphorylation of c-Jun N-terminal kinase (JNK) [90]. It indicates that the regulation of JNK phosphorylation plays a central role in GITR-induced changes in Tregs with therapeutic implications and that JNK inhibitors can act as another regulator of GITR-mediated lung cancer immunotherapy.

The CD39 ectoenzyme produces AMP from ATP or ADP, which is subsequently converted into extracellular adenosine by the CD73 ectoenzyme [91]. Humans can express low levels of adenosine deaminase, while T effector cells are enriched in adenosine deaminase with low levels of CD39 and CD73. It may influence Tregs to produce and sustain high concentrations of extracellular adenosine [92]. Adenosine binding to adenosine A2A receptor (ADORA2A) on activated T effector cells or natural killer (NK) cells generates an immunosuppressive effect through the coordinated expression of CD39/CD73 as one of the mechanisms of Treg-mediated immunosuppression [93]. The ectonucleotidases CD39/CD73 and ADORA2A appear as possible targets for novel treatments in cancer [94]. In addition, exosomes from other cancer cells could express CD39/CD73 to suppress T cells through adenosine production [95]. It is needed to furthermore clarify the expression of CD39 and/or CD73 on exosomes from lung cancer tissues and the axis of CD39/CD73 and ADORA2A as a potential target for inhibition of Treg-mediated immunosuppression and lung cancer immunotherapy. CD95 (APO-1, Fas) is a member of the death receptor family and a subfamily of the tumor necrosis factor receptor superfamily. Activation-induced cell death is involved in the removal of activated T cells and Ag-presenting B cells depending, at least partially, on death receptor CD95 and the ligand CD95L [96, 97]. Human Tregs in lung cancer could express Fas and FasL [98] and low levels of CD95L upon stimulation, inducing the resistance to activation-induced cell death in Tregs. The exogenous CD95 stimulation by administration of an agonistic anti-CD95 antibody reduced systemic numbers of Tregs and Foxp3+GFP+ Treg numbers in pooled lymph nodes. Chen et al. [99] engineered cancer cells with CD95L to kill Tregs within the tumor, leading to a decrease in tumor mass. CD95L-expressing conventional CD4+ T in the tumor can reduce the number of Tregs [100]. Tregs were depleted from the inflamed tissue site by an unknown cell type expressing CD95L [101]. It implies that Tregs are susceptible to homeostatic controls by exogenous CD95 stimulation, which provides a new potential target for Treg-targeting lung cancer immunotherapy. The perforin/granzyme pathway is a central component of antitumor and antiviral effector immune responses mediated by cytotoxic T lymphocytes and NK cells. Granzyme B and perforin were also involved in the suppression of nodal metastasis of lung cancer cells [102]. Tregs suppressed the ability of NK cells and cytotoxic T lymphocytes to clear tumor cells via granzymes A/B/perforin within Tregs [103]. Nonregulatory responder CD4+ T cells could resist Treg suppression by production of granzyme B [104]. A recent study suggests that lung cancer cells utilize the increased PI-9 (granzyme B inhibitor) expression to protect from granzyme Bmediated cytotoxicity as an immune evasion mechanism of lung cancer [105]. Czystowska et al. [106] further

Cancer Metastasis Rev

demonstrated that human CD4 + CD25 + Tregs and CD4+CD25− T cells reciprocally regulate death/growth arrest by differentially utilizing the granzyme–perforin pathway depending on IL-2 concentrations. It seems that the roles of the granzyme–perforin pathway in lung cancer immunity are complicated, probably depending on the microenvironment. LL-37, a human cathelicidin, can kill stimulated and nonstimulated CD4+CD25+FoxP3+ Tregs through apoptosis, rather than CD4+CD25− T cells [107]. LL-37-induced apoptosis in Tregs depends upon granzymes A and B from cytolytic granules. Administration of LL-37 within the tumor could influence the adaptive antitumor immune response by killing Tregs and, thus, inhibiting the suppressor activity. The role of LL-37 in lung cancer immunotherapy needs to be furthermore clarified. Nrp-1 expressed on the surface of CD4+CD25+ Tregs independent of the activation can be downregulated in naive CD4+CD25− T cells after the stimulation [108]. Nrp-1 is expressed by most Tregs rather than naive T helper cells and promotes prolonged interactions with immature DCs [109]. Nrp-1 is also a high-affinity receptor for latent and active TGF-β1, activates the latent form, and is relevant to Treg activity in tumor biology [110]. Nrp1+ Tregs showed to be more efficient to induce the suppression, and Nrp1+ and Nrp1− Tregs decreased in tumor-draining lymph nodes of patients with cervical cancer following preoperative chemoradiotherapy [111]. Nrp1+ Treg elimination may facilitate the generation of T cells to mediate the destruction of the neoplastic cells after cytotoxic therapy. Recently, Nrp-1 was identified as a valid antiangiogenic target in multimalignancy, including lung cancer, and as a potential direct antitumor target

Fig. 3 Role of TGF-β in lung cancer pathogenesis and progression. TGF-β within lung cancer microenvironment upregulates B7H1 and GITRL expression in DCs and de novo generation and expansion of Tregs. TGF-β induces epithelial-to-mesenchymal-like transition (EMT), associated with early NSCLC progression, while the process can be inhibited by BMP7 and MiR-23a. TGF-β induces expression of FOXP3 in lung cancer cells. TGF-β induces lung cancer cells to death

of immunotherapy and antiangiogenic therapy for lung cancer [112].

4.2 Soluble factor-mediated suppression of Tregs Major immunosuppressive cytokines or factors of Tregs include TGF-β, IL-10, IL-35, galectin-1, or prostaglandin E2 (PGE2), as explained in Fig. 3. TGF-β can regulate the biological function of CD4+ T cells and demethylate FOXP3 promoter in CD4+CD25− T cells to maintain lymphocyte homeostasis and increase FOXP3 expression [113]. TGF-β within the lung cancer microenvironment could upregulate PD-L1 and GITRL expression in DC and Treg generation [114]. TGF-β plays a role in tumorigenesis and highly expresses in the lungs at risk for lung cancer [115, 116]. The expression of FOXP3 within tumor cells is associated with tumor progression and metastasis, as a poor prognostic factor [117]. Treatment of TGF-β2 and DNA methyltransferase inhibitor could upregulate FOXP3 expression in lung cancer cells [118]. It is possible to have TGF-β as a target for lung cancer immunotherapy, although the role of TGF-β signaling in tumorigenesis needs to be further clarified [118]. The epithelial-to-mesenchymal-like transition associated with early progression of NSCLC can be induced by TGF-β [119]. However, the loss of TGF-β-induced tumor suppressor function due to genetic and epigenetic alterations of TβRII may play a critical role in lung carcinogenesis [120]. The downregulation of TβRII in lung cancer depended on histone deacetylase and can be restored by the inhibitors in lung cancer patients [121]. It is necessary to balance TGF-β-induced tumor suppression and pathogenesis when TGF-β in the

by binding to TβRII on lung cancer cells. TGF-β transforming growth factor-β, Tregs regulatory T cells, DCs dendritic cells, CTLs cytotoxic T lymphocytes, GITRL glucocorticoid-induced tumor necrosis factor receptor ligand, EMT epithelial-to-mesenchymal-like transition, NSCLC nonsmall cell lung cancer, BMP7 bone morphogenetic protein 7, MiR23 microRNA 23a, TβRII TGF-β type II receptor

Cancer Metastasis Rev

microenvironments can be targeted by immunotherapy for lung cancer. IL-10 is commonly regarded as an anti-inflammatory and immunosuppressive cytokine, allowing tumor immune escape (Fig. 4). It was evidenced that human lung cancer cells could induce IL-10 production from T cells and increase the capacity of T-cell-derived IL-10, associated with clinical prognosis of patients with NSCLC [122, 123]. Polymorphisms of IL-10 genes were associated with the occurrence of NSCLC [124]. NSCLC origin soluble mediators may play an immunoregulatory role through induction of IL-10 production [125]. On the other hand, IL-10 might also possess anticancer properties due to the immunostimulating and antiangiogenic functions [126] and is associated with worse prognosis in NSCLC

[127]. The survival advantage of patients with stage I NSCLC may be related to the antitumor activity of the CD8+/IL-10+ cell phenotype [128]. IL-35 is identified as a novel immunosuppressive/antiinflammatory cytokine, of which two subunits, IL-12A and Epstein-Barr virus induced 3, are produced by Tregs [129, 130]. IL-35 could suppress T-cell proliferation and convert naive T cells into IL-35-producing induced regulatory T cells to mediate the suppression (Fig. 5) [131]. IL-35 is signaled through a unique heterodimer of receptor chains IL-12Rβ2 and gp130 or homodimers of each or both chains, dependent upon the activation of transcription factors STAT1 and STAT4, to bound to distinct sites in promoters of genes encoding p35 and Epstein-Barr virus induced 3 [132]. IL35-producing induced regulatory T cells can be induced in an IL-35- and/or IL-10-dependent manner within the tumor microenvironment to contribute to Treg-mediated tumor progression [131]. Galectin-1 (Gal-1) is a homodimer of noncovalently associated 14 kDa subunits with two carbohydrate recognition domains to enable cell adhesion and cross-linking of several glycoproteins with branched or repeating Galβ1-4GlcNAc sequences on T cells [133]. Gal-1 overexpressed on Tregs and blockade of Gal-1 binding reduced the inhibitory effects of CD4+CD25+ T cells [134]. The primary roles of Gal-1 in cancer progression and metastasis are attributed to suppress Tcell immune responses, promote tumor angiogenesis, and increase tumor cell adhesion and invasion, including lung cancer [135]. Gal-1 might locally contribute to the apoptotic elimination of infiltrating effector T cells and favor tumor progression [136]. Blockage of Gal-1 expression stimulated the

Fig. 5 Role of IL-35 in a tumor microenvironment and IL-35 signaling pathway. IL-35 has two subunits, IL-12A and Epstein-Barr virus induced 3 (EBi3). IL-35 suppresses T-cell proliferation and converts naive T cells and Tconv cells into IL-35-producing induced regulatory T cells (iTr35 cells). The generation of iTr35 cells within cancer can be induced in an IL-35- or IL-10-dependent manner. IL-35 signaled through IL-12Rβ2

and gp130 required for IL-35 expression and conversion into iTr35 cells. The signaling through IL-35 receptor depends on transcription factors STAT1 and STAT4. iTr35s IL-35-producing regulatory T cells, Tconv conventional T cell, EBi3 Epstein-Barr virus induced 3, B16 B16 melanoma cells, MC38 colorectal adenocarcinoma cells, IL-35 interleukin-35, IL-35R IL-35 receptor, IL-2A interleukin-2A

Fig. 4 Role of IL-10 in lung cancer. Lung cancer cells can produce and also induce T cell to produce IL-10, partially depending on PGE2. IL-10 can impair antitumor immunity, due to defects in T-cell and DC function. IL-10 might possess anticancer properties on the basis of immunostimulating and antiangiogenic functions. Tr1 IL-10-secreting T regulatory, DCs dendritic cells

Cancer Metastasis Rev

Fig. 6 Role of galectin-1 in Tregs and lung cancer microenvironment and signaling pathway of galectin-1 (Gal-1)-induced T-cell apoptosis. Gal-1 overexpresses on Tregs, upregulates upon the activation, and overexpresses on cancer cells. Primary roles of Gal-1 in cancer progression and metastasis are through inducing T-cell apoptosis. Signaling pathways of Gal-1-induced T-cell apoptosis are delineated.

Tregs regulatory T cells, DCs dendritic cells, Gal-1 galectin-1, Txnip thioredoxin-interacting protein, ASK1 apoptosis signal-regulating kinase 1, MAPK mitogen-activated protein kinase, JNK c-Jun amino-terminal kinase, AP-1 activator protein-1, ER endoplasmic reticulum, TRE TPA responsive element, Bcl-2 B-cell lymphoma-2, ZAP70 zeta-chainassociated protein kinase

generation of a tumor-specific T-cell-mediated response and tumor rejection [137] and reduced lung metastasis with increased CD4+ and CD8+ T cells and cancer cell adherence [138]. Gal-1 could induce the expression of IL-10 in monocytes and monocyte-derived DCs [139]. It suggests that the Gal-1/IL-10 functional axis may be crucial in lung cancermediated immune suppression. It is evidenced that the JNK/ c-Jun/AP-1 pathway with other T-cell apoptosis pathways plays a key role for T-cell death regulation in response to Gal-1 stimulation [140], as shown in Fig. 6.

PGE2 was found to promote the development of Tregs [141], of which cyclooxygenase (COX)-2-dependent production contributes to TGF-β-induced Treg function in human NSCLC [142, 143]. It seems that TGF-β and PGE2 signaling may be important in therapeutic interventions in lung cancer. Figure 7 demonstrates that COX2 and PGE2 are essential for the EP2- and EP4-dependent induction of Tregs in cancer, interaction of DCs with Tregs, or the suppressive activity of Tregs [144]. A recent study demonstrated that COX2 -1195G/ A polymorphism could influence the infiltration of Tregs into

Fig. 7 Role of PGE2 in Treg-mediated antitumor immunosuppression and lung cancer microenvironment. PGE 2 are small molecule derivatives of arachidonic acid (AA), produced by cyclooxygenases (COX; COX 1 /COX 2 ) and PG synthases. It promotes Treg differentiation from naive T cell and expansion of re-existing Tregs. PGE2 can mediate the suppressive activity of Tregs. PGE2 selectively

suppresses effector functions of the Th1-, CTL-, and NK cell-mediated type 1 immunity and supports activation of DCs. PGE2 contributes cell proliferation and migration of lung cancer. Tregs regulatory T cells, DCs dendritic cells, AA arachidonic acid, COX2 cyclooxygenase-2, PG synthases prostaglandin synthases, PGE2 prostaglandin E2, TGF-β transforming growth factor-β

Cancer Metastasis Rev

NSCLC which was reflected by COX2 SNP factor as a prognostic factor [145]. PGE2, in an autocrine or paracrine fashion, modulates transcriptional repressors of E-cadherin and regulates COX2-dependent E-cadherin expression in NSCLC [146, 147]. It suggests that the blocking of PGE2 production or activity by genetic or pharmacological interventions may benefit patients with NSCLC. The possible link between tumor COX2 overexpression and elevated Erk-mediated cancer cell proliferation and migration may contribute to EGFR inhibitor resistance in NSCLC [148]. Increased expression of PGE2 in the lung tumor microenvironment may initiate a mitogen-signaling cascade composed of EP4, βArrestin1, and c-Src, to mediate lung cancer cell migration [149]. Selective targeting of EP4 with a ligand antagonist may provide an efficient approach to better manage patients with advanced lung cancer. Changes in urine PGE-M, a major PGE2 metabolite, reflect the levels of systemic and intratumoral PGE2. A phase II clinical trial demonstrated that the more than 70 % decline in PGE-M was associated with an apparent survival benefit in NSCLC patients [150].

Authors’ contributions DZ contributed to the collection of data, discussion, writing, and review of figures, and ZHC, DCW, and XDW contributed to the revision, structure design, manuscript preparation, and discussion. Conflict of interest The authors declare no conflict of interest.

References 1.

2. 3.

4.

5.

5 Conclusion The present article gives an overview of the biological functions and regulations of Tregs associated with the development of lung cancer and metastasis and explores the potentials of Treg-oriented therapeutic targets. Subsets and features of Tregs mainly include naturally occurring Tregs (CD4+ nTregs and CD8+ nTregs) and adaptive/induced Tregs (CD4+ iTregs and CD8+ iTregs). Tregs, especially in circulation or regional lymph nodes, play an important role in the progress and metastasis of lung cancer and are considered as therapeutic targets and biomarkers to predict the survival length and recurrence of lung cancer. Tregs may suppress the function of immune cells and allow lung cancer escapes probably through cell interaction-dependent or cytokine-mediated suppression, although the exact mechanism of Treg cell-mediated suppression remains unclear. Increasing understanding of Tregs’ functional mechanisms will lead to a number of clinical trials on the discovery and development of Treg-oriented new therapies. Tregs as one of the therapeutic target candidates for lung cancer need global forces and databases to integrate the genes, proteins, receptors, signal pathways, and functions with clinical informatics and phenotypes together.

6. 7.

8.

9.

10.

11.

12. 13.

14. Acknowledgments The work was supported by Zhongshan Distinguished Professor Grant (XDW), The National Nature Science Foundation of China (91230204, 81270099, 81320108001, 81270131, 81300010), The Shanghai Committee of Science and Technology (12JC1402200, 12431900207, 11410708600, 14431905100), Operation Funding of Shanghai Institute of Clinical Bioinformatics, and Ministry of Education, Academic Special Science and Research Foundation for PhD Education (20130071110043).

15.

Torre, L. A., Bray, F., Siegel, R. L., Ferlay, J., Lortet‐Tieulent, J., & Jemal, A. (2015). Global cancer statistics, 2012. CA: A Cancer Journal for Clinicians, 65(2), 87–108. Siegel, R. L., Miller, K. D., & Jemal, A. (2015). Cancer statistics, 2015. CA: A Cancer Journal for Clinicians, 65(1), 5–29. Schwartz Albiez, R., Monteiro, R., Rodriguez, M., Binder, C., & Shoenfeld, Y. (2009). Natural antibodies, intravenous immunoglobulin and their role in autoimmunity, cancer and inflammation. Clinical and Experimental Immunology, 158, 43–50. Moriya, K., Wakabayashi, A., Shimizu, M., Tamura, H., Dan, K., & Takahashi, H. (2010). Induction of tumor-specific acquired immunity against already established tumors by selective stimulation of innate DEC-205 + dendritic cells. Cancer Immunology, Immunotherapy, 59(7), 1083–1095. Nanni, P., Nicoletti, G., Palladini, A., Croci, S., Murgo, A., Antognoli, A., Landuzzi, L., Fabbi, M., Ferrini, S., & Musiani, P. (2007). Antimetastatic activity of a preventive cancer vaccine. Cancer Research, 67(22), 11037–11044. Zou, W. (2006). Regulatory T cells, tumour immunity and immunotherapy. Nature Reviews Immunology, 6(4), 295–307. Kost, S. E., Kakal, J. A., & Nelson, B. H. (2012). The prognostic value of FoxP3+ tumor-infiltrating lymphocytes in cancer: a critical review of the literature. Clinical Cancer Research, 18(11), 3022–3029. Tao, H., Mimura, Y., Aoe, K., Kobayashi, S., Yamamoto, H., Matsuda, E., Okabe, K., Matsumoto, T., Sugi, K., & Ueoka, H. (2012). Prognostic potential of FOXP3 expression in non-small cell lung cancer cells combined with tumor-infiltrating regulatory T cells. Lung Cancer, 75(1), 95–101. Gershon, R. K., & Kondo, K. (1970). Cell interactions in the induction of tolerance: the role of thymic lymphocytes. Immunology, 18(5), 723. Berendt, M. J., & North, R. J. (1980). T-cell-mediated suppression of anti-tumor immunity. An explanation for progressive growth of an immunogenic tumor. The Journal of Experimental Medicine, 151(1), 69. Hori, S., Nomura, T., & Sakaguchi, S. (2003). Control of regulatory T cell development by the transcription factor Foxp3. Science’s STKE, 299(5609), 1057. Bluestone, J. A., & Abbas, A. K. (2003). Natural versus adaptive regulatory T cells. Nature Reviews Immunology, 3(3), 253–257. Bayer A. L., & Malek T. R. (2009). The role of IL-2 in the development and peripheral homeostasis of naturally occurring CD4+ CD25+ Foxp3+ regulatory T cells. Regulatory T Cells and Clinical Application, 1–20 Wing, K., Onishi, Y., Prieto-Martin, P., Yamaguchi, T., Miyara, M., Fehervari, Z., Nomura, T., & Sakaguchi, S. (2008). CTLA-4 control over Foxp3+ regulatory T cell function. Science’s STKE, 322(5899), 271. Bombardieri, M., Alunno, A., Kelly, S., Bistoni, O., Pitzalis, C., Whyte, S., Gerli, R., Nocentini, G., & Riccardi, C. (2011). Glucocorticoid induced TNF receptor related protein (GITR) is a marker of regulatory T cells: relationship with FOXP3 expression in healthy donors and in patients with rheumatoid arthritis before

Cancer Metastasis Rev

16. 17.

18.

19.

20.

21.

22.

23. 24.

25.

26.

27.

28.

29.

30.

31.

32.

and after corticosteroid therapy. Annals of the Rheumatic Diseases, 70, 685–685. Lourenço, E. V., & La Cava, A. (2011). Natural regulatory T cells in autoimmunity. Autoimmunity, 44(1), 33. Liu, W., Putnam, A. L., Xu-Yu, Z., Szot, G. L., Lee, M. R., Zhu, S., Gottlieb, P. A., Kapranov, P., Gingeras, T. R., de St, F., Groth, B., Clayberger, C., Soper, D. M., Ziegler, S. F., & Bluestone, J. A. (2006). CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ Treg cells. The Journal of Experimental Medicine, 203(7), 1701–1711. Miyara, M., Yoshioka, Y., Kitoh, A., Shima, T., Wing, K., Niwa, A., Parizot, C., Taflin, C., Heike, T., & Valeyre, D. (2009). Functional delineation and differentiation dynamics of human CD4 + T cells expressing the FoxP3 transcription factor. Immunity, 30(6), 899–911. Thornton, A. M., Korty, P. E., Tran, D. Q., Wohlfert, E. A., Murray, P. E., Belkaid, Y., & Shevach, E. M. (2010). Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. The Journal of Immunology, 184(7), 3433. Gottschalk, R. A., Corse, E., & Allison, J. P. (2012). Expression of Helios in peripherally induced Foxp3+ regulatory T cells. The Journal of Immunology, 188(3), 976–980. Hippen, K. L., Merkel, S. C., Schirm, D. K., Sieben, C. M., Sumstad, D., Kadidlo, D. M., McKenna, D. H., Bromberg, J. S., Levine, B. L., & Riley, J. L. (2011). Massive ex vivo expansion of human natural regulatory T cells (Tregs) with minimal loss of in vivo functional activity. Science Translational Medicine, 83, 83ra41–83ra41. Shevach, E. M., & Thornton, A. M. (2014). tTregs, pTregs, and iTregs: similarities and differences. Immunological Reviews, 259(1), 88–102. Mills, K. H. G. (2004). Regulatory T cells: friend or foe in immunity to infection? Nature Reviews Immunology, 4(11), 841–855. Battaglia, M., Gregori, S., Bacchetta, R., & Roncarolo, M. G. (2006). Tr1 cells: from discovery to their clinical application. Seminars in Immunology, 18(2), 120–7. Maynard, C. L., Harrington, L. E., Janowski, K. M., Oliver, J. R., Zindl, C. L., Rudensky, A. Y., & Weaver, C. T. (2007). Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3− precursor cells in the absence of interleukin 10. Nature Immunology, 8(9), 931–941. Mandapathil, M., & Whiteside, T. L. (2011). Targeting human inducible regulatory T cells (Tr1) in patients with cancer: blocking of adenosine-prostaglandin E2 cooperation. Expert Opinion on Biological Therapy, 11(9), 1203–1214. Brun, V., Bastian, H., Neveu, V., & Foussat, A. (2009). Clinical grade production of IL-10 producing regulatory Tr1 lymphocytes for cell therapy of chronic inflammatory diseases. International Immunopharmacology, 9(5), 609–613. Santos, L., Al-Sabbagh, A., Londono, A., & Weiner, H. L. (1994). Oral tolerance to myelin basic protein induces regulatory TGF-βsecreting T cells in Peyer’s patches of SJL mice. Cellular Immunology, 157(2), 439–447. Weiner, H. L. (2001). Induction and mechanism of action of transforming growth factor-β-secreting Th3 regulatory cells. Immunological Reviews, 182(1), 207–214. Carrier, Y., Yuan, J., Kuchroo, V. K., & Weiner, H. L. (2007). Th3 cells in peripheral tolerance. I. Induction of Foxp3-positive regulatory T cells by Th3 cells derived from TGF-β T cell-transgenic mice. The Journal of Immunology, 178(1), 179. Curotto de Lafaille, M. A., & Lafaille, J. J. (2009). Natural and adaptive Foxp3+ regulatory T cells: more of the same or a division of labor? Immunity, 30(5), 626–635. Liu, V. C., Wong, L. Y., Jang, T., Shah, A. H., Park, I., Yang, X., Zhang, Q., Lonning, S., Teicher, B. A., & Lee, C. (2007). Tumor

evasion of the immune system by converting CD4+ CD25− T cells into CD4+ CD25+ T regulatory cells: role of tumor-derived TGFβ. The Journal of Immunology, 178(5), 2883. 33. Al‐Qahtani, D., Anil, S., & Rajendran, R. (2011). Tumour infiltrating CD25+ FoxP3+ regulatory T cells (Tregs) relate to tumour grade and stromal inflammation in oral squamous cell carcinoma. Journal of Oral Pathology & Medicine, 40(8), 636–642. 34. Mayer, C. T., Floess, S., Baru, A. M., Lahl, K., Huehn, J., & Sparwasser, T. (2011). CD8+ Foxp3+ T cells share developmental and phenotypic features with classical CD4+ Foxp3+ regulatory T cells but lack potent suppressive activity. European Journal of Immunology, 41(3), 716–725. 35. Wang, R. F. (2008). CD8+ regulatory T cells, their suppressive mechanisms, and regulation in cancer. Human Immunology, 69(11), 811–814. 36. Woo, E. Y., Chu, C. S., Goletz, T. J., Schlienger, K., Yeh, H., Coukos, G., Rubin, S. C., Kaiser, L. R., & June, C. H. (2001). Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer. Cancer Research, 61(12), 4766–4772. 37. Woo, E. Y., Yeh, H., Chu, C. S., Schlienger, K., Carroll, R. G., Riley, J. L., Kaiser, L. R., & June, C. H. (2002). Cutting edge: regulatory T cells from lung cancer patients directly inhibit autologous T cell proliferation. Journal of Immunology, 168(9), 4272– 4276. 38. Okita, R., Saeki, T., Takashima, S., Yamaguchi, Y., & Toge, T. (2005). CD4+CD25+ regulatory T cells in the peripheral blood of patients with breast cancer and non-small cell lung cancer. Oncology Reports, 14(5), 1269–1273. 39. Meloni, F., Morosini, M., Solari, N., Passadore, I., Nascimbene, C., Novo, M., Ferrari, M., Cosentino, M., Marino, F., Pozzi, E., & Fietta, A. M. (2006). Foxp3 expressing CD4+ CD25+ and CD8+CD28− T regulatory cells in the peripheral blood of patients with lung cancer and pleural mesothelioma. Human Immunology, 67(1–2), 1–12. 40. Wang, Y. Y., He, X. Y., Cai, Y. Y., Wang, Z. J., & Lu, S. H. (2011). The variation of CD4+CD25+ regulatory T cells in the periphery blood and tumor microenvironment of non-small cell lung cancer patients and the downregulation effects induced by CpG ODN. Targeted Oncology, 6(3), 147–154. 41. Su, Y. J., Ren, K., Li, H., Ren, X. B., & Wang, C. L. (2007). Clinical significance of CD4+ CD25+ regulatory T-cells detection in tumor-draining lymph nodes of nonsmall cell lung cancer patients. Zhonghua Zhong Liu Za Zhi, 29(12), 922–926. 42. Ju, S., Qiu, H., Zhou, X., Zhu, B., Lv, X., Huang, X., Li, J., Zhang, Y., Liu, L., Ge, Y., Johnson, D. E., & Shu, Y. (2009). CD13+CD4+CD25hi regulatory T cells exhibit higher suppressive function and increase with tumor stage in non-small cell lung cancer patients. Cell Cycle, 8(16), 2578–2585. 43. Karagoz, B., Bilgi, O., Gumus, M., Erikci, A. A., Sayan, O., Turken, O., Kandemir, E. G., Ozturk, A., & Yaylaci, M. (2010). CD8+CD28− cells and CD4+CD25+ regulatory T cells in the peripheral blood of advanced stage lung cancer patients. Medical Oncology, 27(1), 29–33. 44. Koyama, K., Kagamu, H., Miura, S., Hiura, T., Miyabayashi, T., Itoh, R., Kuriyama, H., Tanaka, H., Tanaka, J., Yoshizawa, H., Nakata, K., & Gejyo, F. (2008). Reciprocal CD4+ T-cell balance of effector CD62Llow CD4+ and CD62LhighCD25+ CD4+ regulatory T cells in small cell lung cancer reflects disease stage. Clinical Cancer Research, 14(21), 6770–6779. 45. Wang W., Hodkinson P., McLaren F., Mackinnon A., Wallace W., Howie S., & Sethi T. (2012). Small cell lung cancer tumour cells induce regulatory T lymphocytes, and patient survival correlates negatively with FOXP3(+) cells in tumour infiltrate. Int J Cancer, 15;131(6):E928–37.

Cancer Metastasis Rev 46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

Hasegawa, T., Suzuki, H., Yamaura, T., Muto, S., Okabe, N., Osugi, J., Hoshino, M., Higuchi, M., Ise, K., & Gotoh, M. (2014). Prognostic value of peripheral and local forkhead box P3+ regulatory T cells in patients with non-small-cell lung cancer. Molecular and Clinical Oncology, 2(5), 685–694. Hanagiri, T., Shigematsu, Y., Shinohara, S., Takenaka, M., Oka, S., Chikaishi, Y., Nagata, Y., Iwata, T., Uramoto, H., & So, T. (2013). Clinical significance of the frequency of regulatory T cells in regional lymph node lymphocytes as a prognostic factor for non-small-cell lung cancer. Lung Cancer, 81(3), 475–479. Black, C. C., Turk, M. J., Dragnev, K., & Rigas, J. R. (2013). Adenocarcinoma contains more immune tolerance regulatory Tcell lymphocytes (versus squamous carcinoma) in non-small-cell lung cancer. Lung, 191(3), 265–270. Kinoshita, T., Ishii, G., Hiraoka, N., Hirayama, S., Yamauchi, C., Aokage, K., Hishida, T., Yoshida, J., Nagai, K., & Ochiai, A. (2013). Forkhead box P3 regulatory T cells coexisting with cancer associated fibroblasts are correlated with a poor outcome in lung adenocarcinoma. Cancer Science, 104(4), 409–415. Li, J.-Y., Duan, X.-F., Wang, L.-P., Xu, Y.-J., Huang, L., Zhang, T.-F., Liu, J.-Y., Li, F., Zhang, Z., & Yue, D.-L. (2014). Selective depletion of regulatory T cell subsets by docetaxel treatment in patients with nonsmall cell lung cancer. Journal of Immunology Research, 2014, 286170. doi:10.1155/2014/286170. Teng, M. W. L., Ritchie, D. S., Neeson, P., & Smyth, M. J. (2011). Biology and clinical observations of regulatory T cells in cancer immunology. Cancer Immunology and Immunotherapy, 61–95. Peggs, K. S., Quezada, S. A., Korman, A. J., & Allison, J. P. (2006). Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. Current Opinion in Immunology, 18(2), 206– 213. Keilholz, U. (2008). CTLA-4: negative regulator of the immune response and a target for cancer therapy. Journal of Immunotherapy, 31(5), 431. Ackerman, A., Klein, O., McDermott, D.F., Wang, W., Ibrahim, N., Lawrence, D.P., Gunturi, A., Flaherty, K.T., Hodi, F.S., Kefford, R., Menzies, A.M., Atkins, M.B., Long, G.V., & Sullivan, R.J. (2014). Outcomes of patients with metastatic melanoma treated with immunotherapy prior to or after BRAF inhibitors. Cancer. 1;120(11):1695–701. Erfani, N., Mehrabadi, S. M., Ghayumi, M. A., Haghshenas, M. R., Mojtahedi, Z., Ghaderi, A., & Amani, D. (2012). Increase of regulatory T cells in metastatic stage and CTLA-4 over expression in lymphocytes of patients with non-small cell lung cancer (NSCLC). Lung Cancer, 77(2), 306–11. Karabon, L., Pawlak, E., Tomkiewicz, A., Jedynak, A., Passowicz-Muszynska, E., Zajda, K., Jonkisz, A., Jankowska, R., Krzakowski, M., & Frydecka, I. (2011). CTLA-4, CD28, and ICOS gene polymorphism associations with non-small-cell lung cancer. Human Immunology, 72(10), 947–54. Mellor, A. L., & Munn, D. H. (2004). IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nature Reviews Immunology, 4(10), 762–774. Salvi S., Fontana V., Boccardo S., Merlo D. F., Margallo E., Laurent S., Morabito A., Rijavec E., Dal Bello M. G., & Mora M. (2012). Evaluation of CTLA-4 expression and relevance as a novel prognostic factor in patients with non-small cell lung cancer. Cancer Immunology, Immunotherapy, 1–10. Leach, D. R., Krummel, M. F., & Allison, J. P. (1996). Enhancement of antitumor immunity by CTLA-4 blockade. Science, 271(5256), 1734–1736. Hodi, F. S., O’Day, S. J., McDermott, D. F., Weber, R. W., Sosman, J. A., Haanen, J. B., Gonzalez, R., Robert, C., Schadendorf, D., & Hassel, J. C. (2010). Improved survival with ipilimumab in patients with metastatic melanoma. New England Journal of Medicine, 363(8), 711–723.

61.

Robert, C., Thomas, L., Bondarenko, I., O’Day, S., Weber, J., Garbe, C., Lebbe, C., Baurain, J.-F., Testori, A., & Grob, J.-J. (2011). Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. New England Journal of Medicine, 364(26), 2517–2526. 62. Lynch, T. J., Bondarenko, I., Luft, A., Serwatowski, P., Barlesi, F., Chacko, R., Sebastian, M., Neal, J., Lu, H., & Cuillerot, J.-M. (2012). Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. Journal of Clinical Oncology, 30(17), 2046–2054. 63. Golden, E. B., Demaria, S., Schiff, P. B., Chachoua, A., & Formenti, S. C. (2013). An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunology Research, 1(6), 365–372. 64. Reck, M., Bondarenko, I., Luft, A., Serwatowski, P., Barlesi, F., Chacko, R., Sebastian, M., Lu, H., Cuillerot, J.-M., & Lynch, T. (2013). Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Annals of Oncology, 24(1), 75–83. 65. Kamphorst, A. O., & Ahmed, R. (2013). Manipulating the PD-1 pathway to improve immunity. Current Opinion in Immunology, 25(3), 381–388. 66. Waki, K., Yamada, T., Yoshiyama, K., Terazaki, Y., Sakamoto, S., Matsueda, S., Komatsu, N., Sugawara, S., Takamori, S., & Itoh, K. (2014). PD-1 expression on peripheral blood T-cell subsets correlates with prognosis in non-small cell lung cancer. Cancer Science, 105(10), 1229–1235. 67. Topalian, S. L., Drake, C. G., & Pardoll, D. M. (2012). Targeting the PD-1/B7-H1 (PD-L1) pathway to activate anti-tumor immunity. Current Opinion in Immunology, 24(2), 207–212. 68. Zhang, Y., Huang, S., Gong, D., Qin, Y., & Shen, Q. (2010). Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8+T lymphocytes in human non-small cell lung cancer. Cellular & Molecular Immunology, 7(5), 389–395. 69. Lipson, E. J., Sharfman, W. H., Drake, C. G., Wollner, I., Taube, J. M., Anders, R. A., Xu, H., Yao, S., Pons, A., & Chen, L. (2013). Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clinical Cancer Research, 19(2), 462–468. 70. Gettinger, S., Horn, L., Gandhi, L., Spigel, D., Antonia, S., Rizvi, N., Powderly, J., Heist, R., Carvajal, R., & Jackman, D. (2014). Long-term survival, clinical activity, and safety of nivolumab (anti-PD-1; BMS-936558, ONO-4538) in patients (pts) with advanced non-small cell lung cancer (NSCLC). International Journal of Radiation Oncology Biology Physics, 90(5), S34. 71. Topalian, S. L., Hodi, F. S., Brahmer, J. R., Gettinger, S. N., Smith, D. C., McDermott, D. F., Powderly, J. D., Carvajal, R. D., Sosman, J. A., & Atkins, M. B. (2012). Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. New England Journal of Medicine, 366(26), 2443–2454. 72. Antonia S. J., Gettinger S., Chow L. Q., Juergens R., Borghaei H., Shen Y., Harbison C., Chen A. C., Ready N. E., & Rizvi N. A. (2014). Nivolumab (anti-PD-1; BMS-936558, ONO-4538) and ipilimumab in first-line non-small cell lung cancer (NSCLC): interim phase 1 results. Journal of clinical oncology, 32,5s. 73. Okamura, T., Fujio, K., Sumitomo, S., & Yamamoto, K. (2012). Roles of LAG3 and EGR2 in regulatory T cells. Annals of the Rheumatic Diseases, 71(Suppl 2), i96–i100. 74. Gandhi, M. K., Lambley, E., Duraiswamy, J., Dua, U., Smith, C., Elliott, S., Gill, D., Marlton, P., Seymour, J., & Khanna, R. (2006). Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients. Blood, 108(7), 2280.

Cancer Metastasis Rev 75.

Prigent, P., Dréano, M., & Triebel, F. (1999). Lymphocyte activation gene-3 induces tumor regression and antitumor immune responses. European Journal of Immunology, 29(12), 3867–3876. 76. Brignone, C., Grygar, C., Marcu, M., Perrin, G., & Triebel, F. (2007). IMP321 (sLAG-3) safety and T cell response potentiation using an influenza vaccine as a model antigen: a single-blind phase I study. Vaccine, 25(24), 4641–4650. 77. Brignone, C., Escudier, B., Grygar, C., Marcu, M., & Triebel, F. (2009). A phase I pharmacokinetic and biological correlative study of IMP321, a novel MHC class II agonist, in patients with advanced renal cell carcinoma. Clinical Cancer Research, 15(19), 6225–6231. 78. Brignone, C., Gutierrez, M., Mefti, F., Brain, E., Jarcau, R., Cvitkovic, F., Bousetta, N., Medioni, J., Gligorov, J., & Grygar, C. (2010). First-line chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel and IMP321 (LAG-3Ig) enhances immune responses and antitumor activity. Journal of Translational Medicine, 8(1), 71. 79. Camisaschi, C., Casati, C., Rini, F., Perego, M., De Filippo, A., Triebel, F., Parmiani, G., Belli, F., Rivoltini, L., & Castelli, C. (2010). LAG-3 expression defines a subset of CD4 + CD25highFoxp3+ regulatory T cells that are expanded at tumor sites. The Journal of Immunology, 184(11), 6545–6551. 80. Shimizu, J., Yamazaki, S., Takahashi, T., Ishida, Y., & Sakaguchi, S. (2002). Stimulation of CD25+ CD4+ regulatory T cells through GITR breaks immunological self-tolerance. Nature Immunology, 3(2), 135–142. 81. Cohen, A. D., Schaer, D. A., Liu, C., Li, Y., HirschhornCymmerman, D., Kim, S. C., Diab, A., Rizzuto, G., Duan, F., & Perales, M. A. (2010). Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. Plos One, 5(5), e10436. 82. Zhou, P., L’italien, L., Hodges, D., & Schebye, X. M. (2007). Pivotal roles of CD4+ effector T cells in mediating agonistic anti-GITR mAb-induced-immune activation and tumor immunity in CT26 tumors. The Journal of Immunology, 179(11), 7365– 7375. 83. Houot, R., & Levy, R. (2009). T-cell modulation combined with intratumoral CpG cures lymphoma in a mouse model without the need for chemotherapy. Blood, 113(15), 3546–3552. 84. Cohen, A. D., Diab, A., Perales, M. A., Wolchok, J. D., Rizzuto, G., Merghoub, T., Huggins, D., Liu, C., Turk, M. J., & Restifo, N. P. (2006). Agonist anti-GITR antibody enhances vaccine-induced CD8+ T-cell responses and tumor immunity. Cancer Research, 66(9), 4904. 85. Imai, N., Ikeda, H., Tawara, I., Wang, L., Nishikawa, H., Kato, T., & Shiku, H. (2009). Glucocorticoid-induced tumor necrosis factor receptor stimulation enhances the multifunctionality of adoptively transferred tumor antigen-specific CD8+ T cells with tumor regression. Cancer Science, 100(7), 1317–1325. 86. Mitsui, J., Nishikawa, H., Muraoka, D., Wang, L., Noguchi, T., Sato, E., Kondo, S., Allison, J. P., Sakaguchi, S., & Old, L. J. (2010). Two distinct mechanisms of augmented antitumor activity by modulation of immunostimulatory/inhibitory signals. Clinical Cancer Research, 16(10), 2781. 87. Pruitt, S. K., Boczkowski, D., de Rosa, N., Haley, N. R., Morse, M. A., Tyler, D. S., Dannull, J., & Nair, S. (2011). Enhancement of anti-tumor immunity through local modulation of CTLA-4 and GITR by dendritic cells. European Journal of Immunology, 41(12), 3553–63. 88. Stephens, G. L., McHugh, R. S., Whitters, M. J., Young, D. A., Luxenberg, D., Carreno, B. M., Collins, M., & Shevach, E. M. (2004). Engagement of glucocorticoid-induced TNFR familyrelated receptor on effector T cells by its ligand mediates resistance

to suppression by CD4 + CD25 + T cells. The Journal of Immunology, 173(8), 5008–5020. 89. Côté, A. L., Zhang, P., O’Sullivan, J. A., Jacobs, V. L., Clemis, C. R., Sakaguchi, S., Guevara-Patiño, J. A., & Turk, M. J. (2011). Stimulation of the glucocorticoid-induced TNF receptor familyrelated receptor on CD8 T cells induces protective and highavidity T cell responses to tumor-specific antigens. The Journal of Immunology, 186(1), 275. 90. Joetham, A., Ohnishi, H., Okamoto, M., Takeda, K., Schedel, M., Domenico, J., Dakhama, A., & Gelfand, E. W. (2012). Loss of T regulatory cell suppression following signaling through glucocorticoid-induced tumor necrosis receptor (GITR) is dependent on c-Jun N-terminal kinase activation. Journal of Biological Chemistry, 287(21), 17100–17108. 91. Haskó, G., Linden, J., Cronstein, B., & Pacher, P. (2008). Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nature Reviews Drug Discovery, 7(9), 759– 770. 92. Mandapathil, M., Hilldorfer, B., Szczepanski, M. J., Czystowska, M., Szajnik, M., Ren, J., Lang, S., Jackson, E. K., Gorelik, E., & Whiteside, T. L. (2010). Generation and accumulation of immunosuppressive adenosine by human CD4+ CD25highFOXP3+ regulatory T cells. Journal of Biological Chemistry, 285(10), 7176. 93. Sitkovsky, M., Lukashev, D., Deaglio, S., Dwyer, K., Robson, S., & Ohta, A. (2008). Adenosine A2A receptor antagonists: blockade of adenosinergic effects and T regulatory cells. British Journal of Pharmacology, 153(S1), S457–S464. 94. Häusler S. F. M., Montalbán del Barrio I., Strohschein J., Anoop Chandran P., Engel J. B., Hönig A., Ossadnik M., Horn E., Fischer B., & Krockenberger M. (2011). Ectonucleotidases CD39 and CD73 on OvCA cells are potent adenosine-generating enzymes responsible for adenosine receptor 2A-dependent suppression of T cell function and NK cell cytotoxicity. Cancer Immunology, Immunotherapy, 1–14. 95. Clayton, A., Al-Taei, S., Webber, J., Mason, M. D., & Tabi, Z. (2011). Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production. The Journal of Immunology, 187(2), 676. 96. Hughes, P. D., Belz, G. T., Fortner, K. A., Budd, R. C., Strasser, A., & Bouillet, P. (2008). Apoptosis regulators Fas and Bim cooperate in shutdown of chronic immune responses and prevention of autoimmunity. Immunity, 28(2), 197–205. 97. Janssens, W., Carlier, V., Wu, B., VanderElst, L., Jacquemin, M. G., & Saint-Remy, J. M. R. (2003). CD4+ CD25+ T cells lyse antigen-presenting B cells by Fas-Fas ligand interaction in an epitope-specific manner. The Journal of Immunology, 171(9), 4604. 98. Strauss, L., Bergmann, C., Gooding, W., Johnson, J. T., & Whiteside, T. L. (2007). The frequency and suppressor function of CD4+ CD25highFoxp3+ T cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clinical Cancer Research, 13(21), 6301–6311. 99. Chen, A., Liu, S., Park, D., Kang, Y., & Zheng, G. (2007). Depleting intratumoral CD4+ CD25+ regulatory T cells via FasL protein transfer enhances the therapeutic efficacy of adoptive T cell transfer. Cancer Research, 67(3), 1291. 100. Gritzapis, A. D., Voutsas, I. F., Lekka, E., Papamichail, M., & Baxevanis, C. N. (2010). Peptide vaccination breaks tolerance to HER-2/neu by generating vaccine-specific FasL+ CD4+ T cells: first evidence for intratumor apoptotic regulatory T cells. Cancer Research, 70(7), 2686. 101. Reardon, C., Wang, A., & McKay, D. M. (2008). Transient local depletion of Foxp3+ regulatory T cells during recovery from colitis via Fas/Fas ligand-induced death. The Journal of Immunology, 180(12), 8316–8326.

Cancer Metastasis Rev 102.

103.

104.

105.

106.

107.

108.

109. 110.

111.

112.

113.

114.

115.

116.

117.

Kontani, K., Sawai, S., Hanaoka, J., Tezuka, N., Inoue, S., & Fujino, S. (2001). Involvement of granzyme B and perforin in suppressing nodal metastasis of cancer cells in breast and lung cancers. European Journal of Surgical Oncology, 27(2), 180–186. Cao, X., Fehniger, T. A., Cai, S. F., & Ley, T. J. (2006). The unique roles of murine granzymes (Gzm) A and B for NK-dependent tumor cell killing and regulatory T cell control of NK cells. Proceedings of the American Association for Cancer Research, 2006(1), 149. Ashley, C. W., & Baecher-Allan, C. (2009). Cutting edge: responder T cells regulate human DR+ effector regulatory T cell activity via granzyme B. The Journal of Immunology, 183(8), 4843–4847. Soriano, C., Mukaro, V., Hodge, G., Ahern, J., Holmes, M., Jersmann, H., Moffat, D., Meredith, D., Jurisevic, C., & Reynolds, P. N. (2012). Increased proteinase inhibitor-9 (PI-9) and reduced granzyme B in lung cancer: mechanism for immune evasion? Lung Cancer, 77(1), 38–45. Czystowska, M., Strauss, L., Bergmann, C., Szajnik, M., Rabinowich, H., & Whiteside, T. L. (2010). Reciprocal granzyme/perforin-mediated death of human regulatory and responder T cells is regulated by interleukin-2 (IL-2). Journal of Molecular Medicine, 88(6), 577–588. Mader, J. S., Ewen, C., Hancock, R. E. W., & Bleackley, R. C. (2011). The human cathelicidin, LL-37, induces granzymemediated apoptosis in regulatory T cells. Journal of Immunotherapy, 34(3), 229. Bruder, D., Probst‐Kepper, M., Westendorf, A. M., Geffers, R., Beissert, S., Loser, K., von Boehmer, H., Buer, J., & Hansen, W. (2004). Frontline: neuropilin-1: a surface marker of regulatory T cells. European Journal of Immunology, 34(3), 623–630. Mizui, M., & Kikutani, H. (2008). Neuropilin-1: the glue between regulatory T cells and dendritic cells? Immunity, 28(3), 302–303. Glinka, Y., & Prud’homme, G. J. (2008). Neuropilin-1 is a receptor for transforming growth factor β-1, activates its latent form, and promotes regulatory T cell activity. Journal of Leukocyte Biology, 84(1), 302–310. Battaglia, A., Buzzonetti, A., Monego, G., Peri, L., Ferrandina, G., Fanfani, F., Scambia, G., & Fattorossi, A. (2008). Neuropilin-1 expression identifies a subset of regulatory T cells in human lymph nodes that is modulated by preoperative chemoradiation therapy in cervical cancer. Immunology, 123(1), 129–138. Jubb, A. M., Strickland, L. A., Liu, S. D., Mak, J., Schmidt, M., & Koeppen, H. (2012). Neuropilin-1 expression in cancer and development. The Journal of Pathology, 226(1), 50–60. Pyzik, M., & Piccirillo, C. (2007). TGF-ß1 modulates Foxp3 expression and regulatory activity in distinct CD4+ T cell subsets. Journal of Leukocyte Biology, 82(2), 335–346. Ni, X., Sui, H., Liu, Y., Ke, S., Wang, Y., & Gao, F. (2012). TGF-β of lung cancer microenvironment upregulates B7H1 and GITRL expression in dendritic cells and is associated with regulatory T cell generation. Oncology Reports, 28(2), 615. Roman, C. D., Morrow, J., Whitehead, R., & Beauchamp, R. D. (2002). Induction of cyclooxygenase-2 and invasiveness by transforming growth factor-β 1 in immortalized mouse colonocytes expressing oncogenic ras. Journal of Gastrointestinal Surgery, 6(3), 304–309. Takizawa, H., Tanaka, M., Takami, K., Ohtoshi, T., Ito, K., Satoh, M., Okada, Y., Yamasawa, F., Nakahara, K., & Umeda, A. (2001). Increased expression of transforming growth factor-β 1 in small airway epithelium from tobacco smokers and patients with chronic obstructive pulmonary disease (COPD). American Journal of Respiratory and Critical Care Medicine, 163(6), 1476–1483. Uva, V., Sfondrini, L., Triulzi, T., Casalini, P., Tagliabue, E., & Balsari, A. (2015). FOXP3 expression in tumor cells and its role in cancer progression. Cancer Research, 75(8), 1703–13.

118.

119.

120.

121.

122.

123.

124.

125.

126.

127.

128.

129.

130.

131.

132.

Um, S. W., Lee, S. H., Kim, H., Kwon, O. J., Kang, J. S., & Lee, W. J. (2011). The regulation of FOXP3 expression by the treatment of TGF-β and the modification of DNA methylation in lung cancer cell lines. Tuberculosis and Respiratory Diseases, 70(3), 206–217. Marconi, P., Patel, K., Thimothy, L., Buchanan, S., Liptay, M., Coon, J., Bonomi, P., & Borgia, J. (2010). Modulation of the epithelial-to-mesenchymal-like transition by BMP7 and TGF-β in non-small cell lung cancer cell lines in vitro. Journal of Clinical Oncology, 28(15), e21016. Cao, M., Seike, M., Soeno, C., Mizutani, H., Kitamura, K., Minegishi, Y., Noro, R., Yoshimura, A., Cai, L., & Gemma, A. (2012). MiR-23a regulates TGF-β-induced epithelialmesenchymal transition by targeting E-cadherin in lung cancer cells. International Journal of Oncology, 41(3), 869–75. Halder, S. K., Cho, Y. J., Datta, A., Anumanthan, G., Ham, A. J. L., Carbone, D. P., & Datta, P. K. (2011). Elucidating the mechanism of regulation of transforming growth factor β type II receptor expression in human lung cancer cell lines. Neoplasia, 13(10), 912. Sharma, S., Stolina, M., Lin, Y., Gardner, B., Miller, P. W., Kronenberg, M., & Dubinett, S. M. (1999). T cell-derived IL-10 promotes lung cancer growth by suppressing both T cell and APC function. The Journal of Immunology, 163(9), 5020. Hatanaka, H., Abe, Y., Kamiya, T., Morino, F., Nagata, J., Tokunaga, T., Oshika, Y., Suemizu, H., Kijima, H., & Tsuchida, T. (2000). Clinical implications of interleukin (IL)-10 induced by non-small-cell lung cancer. Annals of Oncology, 11(7), 815–819. Shih, C. M., Lee, Y. L., Chiou, H. L., Hsu, W. F., Chen, W. E., Chou, M. C., & Lin, L. Y. (2005). The involvement of genetic polymorphism of IL-10 promoter in non-small cell lung cancer. Lung Cancer, 50(3), 291–297. Huang, M., Sharma, S., Mao, J. T., & Dubinett, S. M. (1996). Non-small cell lung cancer-derived soluble mediators and prostaglandin E2 enhance peripheral blood lymphocyte IL-10 transcription and protein production. The Journal of Immunology, 157(12), 5512–5520. Mocellin, S., Marincola, F. M., & Young, H. A. (2005). Interleukin-10 and the immune response against cancer: a counterpoint. Journal of Leukocyte Biology, 78(5), 1043–1051. Soria, J. C., Moon, C., Kemp, B. L., Liu, D. D., Feng, L., Tang, X., Chang, Y. S., Mao, L., & Khuri, F. R. (2003). Lack of interleukin10 expression could predict poor outcome in patients with stage I non-small cell lung cancer. Clinical Cancer Research, 9(5), 1785– 1791. Miotto, D., Cascio, N. L., Stendardo, M., Querzoli, P., Pedriali, M., De Rosa, E., Fabbri, L., Mapp, C., & Boschetto, P. (2010). CD8+ T cells expressing IL-10 are associated with a favourable prognosis in lung cancer. Lung Cancer, 69(3), 355–360. Castellani, M., Anogeianaki, A., Felaco, P., Toniato, E., De Lutiis, M., Shaik, B., Fulcheri, M., Vecchiet, J., Tetè, S., & Salini, V. (2010). IL-35, an anti-inflammatory cytokine which expands CD4+ CD25+ Treg cells. Journal of Biological Regulators and Homeostatic Agents, 24(2), 131. Bardel, E., Larousserie, F., Charlot-Rabiega, P., CoulombL’Herminé, A., & Devergne, O. (2008). Human CD4+ CD25+ Foxp3+ regulatory T cells do not constitutively express IL-35. The Journal of Immunology, 181(10), 6898–6905. Collison, L. W., Pillai, M. R., Chaturvedi, V., & Vignali, D. A. A. (2009). Regulatory T cell suppression is potentiated by target T cells in a cell contact, IL-35- and IL-10-dependent manner. The Journal of Immunology, 182(10), 6121–6128. Collison, L. W., Chaturvedi, V., Henderson, A. L., Giacomin, P. R., Guy, C., Bankoti, J., Finkelstein, D., Forbes, K., Workman, C. J., & Brown, S. A. (2010). IL-35-mediated induction of a potent

Cancer Metastasis Rev regulatory T cell population. Nature Immunology, 11(12), 1093– 1101. 133. Koh, H. S., Lee, C., Lee, K. S., Ham, C. S., Seong, R. H., Kim, S. S., & Jeon, S. H. (2008). CD7 expression and galectin-1-induced apoptosis of immature thymocytes are directly regulated by NFκB upon T-cell activation. Biochemical and Biophysical Research Communications, 370(1), 149–153. 134. Miller, M. C., Nesmelova, I. V., Platt, D., Klyosov, A., & Mayo, K. H. (2009). The carbohydrate-binding domain on galectin-1 is more extensive for a complex glycan than for simple saccharides: implications for galectin–glycan interactions at the cell surface. Biochemical Journal, 421(Pt 2), 211. 135. Wang, J., Lu, Z. H., Gabius, H. J., Rohowsky-Kochan, C., Ledeen, R. W., & Wu, G. (2009). Cross-linking of GM1 ganglioside by galectin-1 mediates regulatory T cell activity involving TRPC5 channel activation: possible role in suppressing experimental autoimmune encephalomyelitis. The Journal of Immunology, 182(7), 4036–4045. 136. Blaskó, A., Fajka-Boja, R., Ion, G., & Monostori, É. (2011). How does it act when soluble? Critical evaluation of mechanism of galectin-1 induced T-cell apoptosis. Acta Biologica Hungarica, 62(1), 106–111. 137. Liu, F. T., & Rabinovich, G. A. (2005). Galectins as modulators of tumour progression. Nature Reviews Cancer, 5(1), 29–41. 138. Ito K., & Ralph S. J. (2012). Inhibiting galectin-1 reduces murine lung metastasis with increased CD4+ and CD8+ T cells and reduced cancer cell adherence. Clinical and Experimental Metastasis, 1–12. 139. Kuo, P.-L., Hung, J.-Y., Huang, S.-K., Chou, S.-H., Cheng, D.-E., Jong, Y.-J., Hung, C.-H., Yang, C.-J., Tsai, Y.-M., & Hsu, Y.-L. (2011). Lung cancer-derived galectin-1 mediates dendritic cell anergy through inhibitor of DNA binding 3/IL-10 signaling pathway. The Journal of Immunology, 186(3), 1521–1530. 140. Brandt, B., Abou-Eladab, E., Tiedge, M., & Walzel, H. (2010). Role of the JNK/c-Jun/AP-1 signaling pathway in galectin-1induced T-cell death. Cell Death & Disease, 1(2), e23. 141. Baratelli, F., Lin, Y., Zhu, L., Yang, S.-C., Heuzé-Vourc’h, N., Zeng, G., Reckamp, K., Dohadwala, M., Sharma, S., & Dubinett, S. M. (2005). Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells. The Journal of Immunology, 175(3), 1483–1490. 142. Sharma, S., Yang, S.-C., Zhu, L., Reckamp, K., Gardner, B., Baratelli, F., Huang, M., Batra, R. K., & Dubinett, S. M. (2005). Tumor cyclooxygenase-2/prostaglandin E2-dependent promotion

of FOXP3 expression and CD4+ CD25+ T regulatory cell activities in lung cancer. Cancer Research, 65(12), 5211–5220. 143. Baratelli, F., Lee, J. M., Hazra, S., Lin, Y., Walser, T. C., Schaue, D., Pak, P. S., Elashoff, D., Reckamp, K., & Zhang, L. (2010). PGE2 contributes to TGF-β induced T regulatory cell function in human non-small cell lung cancer. American Journal of Translational Research, 2(4), 356. 144. Soontrapa, K., Honda, T., Sakata, D., Yao, C., Hirata, T., Hori, S., Matsuoka, T., Kita, Y., Shimizu, T., & Kabashima, K. (2011). Prostaglandin E2–prostaglandin E receptor subtype 4 (EP4) signaling mediates UV irradiation-induced systemic immunosuppression. Proceedings of the National Academy of Sciences, 108(16), 6668–6673. 145. Yukawa, T., Shimizu, K., Maeda, A., Yasuda, K., Saisho, S., Okita, R., & Nakata, M. (2015). Cyclooxygenase-2 genetic variants influence intratumoral infiltration of Foxp3-positive regulatory T cells in non-small cell lung cancer. Oncology Reports, 33(1), 74–80. 146. Dohadwala, M., Yang, S.-C., Luo, J., Sharma, S., Batra, R. K., Huang, M., Lin, Y., Goodglick, L., Krysan, K., & Fishbein, M. C. (2006). Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E2 induces transcriptional repressors ZEB1 and Snail in non-small cell lung cancer. Cancer Research, 66(10), 5338–5345. 147. Dohadwala, M., Batra, R. K., Luo, J., Lin, Y., Krysan, K., Põld, M., Sharma, S., & Dubinett, S. M. (2002). Autocrine/paracrine prostaglandin E2 production by non-small cell lung cancer cells r e g u l a t e s m a t ri x m e t a l l o p r o t e i n a s e- 2 a n d C D 4 4 i n cyclooxygenase-2-dependent invasion. Journal of Biological Chemistry, 277(52), 50828–50833. 148. Krysan, K., Reckamp, K. L., Dalwadi, H., Sharma, S., Rozengurt, E., Dohadwala, M., & Dubinett, S. M. (2005). Prostaglandin E2 activates mitogen-activated protein kinase/Erk pathway signaling and cell proliferation in non-small cell lung cancer cells in an epidermal growth factor receptor-independent manner. Cancer Research, 65(14), 6275–6281. 149. Kim, J. I., Lakshmikanthan, V., Frilot, N., & Daaka, Y. (2010). Prostaglandin E2 promotes lung cancer cell migration via EP4βArrestin1-c-Src signalsome. Molecular Cancer Research, 8(4), 569–577. 150. Horn, L., Milne, G., Sandler, A., Morrow, J., Carbone, D., Shyr, Y., Hayes, A., Campbell, N., & Johnson, D. H. (2009). Urine PGE-M to assess prostaglandin E2 (PGE2) levels in non-small cell lung cancer (NSCLC). Journal of Clinical Oncology, 27, e19026.

Regulatory T cells and potential inmmunotherapeutic targets in lung cancer.

Lung cancer and metastasis are two of the most lethal diseases globally and seldom have effective therapies. Immunotherapy is considered as one of the...
2MB Sizes 3 Downloads 10 Views