1479-5876-11-147 1479-5876 Review <p>Immunity of human epithelial ovarian carcinoma: the paradigm of immune suppression in cancer</p> LavouéVincentvincent.lavoue@gmail.com ThédrezAurélieaurelie.thedrez@gmail.com LevêqueJeanjean.leveque@chu-rennes.fr FoucherFabricefabrice.foucher@chu-rennes.fr HennoSébastiensebastien.henno@chu-rennes.fr JauffretVincentvincent.jauffret@gmail.com Belaud-RotureauMarc-Antoinemarc-antoine.bellaud-rotureau@chu-rennes.fr CatrosVeroniqueveronique.catros@chu-rennes.fr CabillicFlorianflorian.cabillic@chu-rennes.fr

Lady Davis Institut, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada

Faculty of Medicine, University of Rennes 1, Rennes, France

Liver Metabolisms and Cancer, Inserm, UMR991, Rennes, France

Department of gynecologic surgery, Teaching Hospital of Rennes, Rennes, France

Department of pathology, Teaching Hospital of Rennes, Rennes, France

Department of cell biology, Teaching Hospital of Rennes, Rennes, France

Service de chirurgie gynécologique, CHU Hôpital Sud, 16 Bd de Bulgarie, Rennes, 35 000, France

Journal of Translational Medicine
<p>Cancer microenvironment</p>
1479-5876 2013 11 1 147 http://www.translational-medicine.com/content/11/1/147 10.1186/1479-5876-11-147
221201327520131362013 2013Lavoué et al.; licensee BioMed Central Ltd.This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Ovarian carcinoma Immunity Immune suppression

Abstract

Epithelial ovarian cancer (EOC) is a significant cause of cancer-related mortality in women, and there has been no substantial decrease in the death rates due to EOC in the last three decades. Thus, basic knowledge regarding ovarian tumor cell biology is urgently needed to allow the development of innovative treatments for EOC. Traditionally, EOC has not been considered an immunogenic tumor, but there is evidence of an immune response to EOC in patients. Clinical data demonstrate that an antitumor immune response and immune evasion mechanisms are correlated with a better and lower survival, respectively, providing evidence for the immunoediting hypothesis in EOC. This review focuses on the immune response and immune suppression in EOC. The immunological roles of chemotherapy and surgery in EOC are also described. Finally, we detail pilot data supporting the efficiency of immunotherapy in the treatment of EOC and the emerging concept that immunomodulation aimed at counteracting the immunosuppressive microenvironment must be associated with immunotherapy strategies.

Introduction

Epithelial ovarian cancer (EOC) is the fifth most common cancer among women and the fourth most common cause of cancer-related death among women in developing countries 1 . The prognosis is poor, with a 5-year survival rate of 30%. The majority of patients relapse within 16–18 months following the end of treatment and die from the disease despite response to first-line therapy consisting of debulking surgery and chemotherapy 2 3 . 15% of patients die within the first year. No substantial decrease in the death rate occurred in the last three decades. Thus, there is an urgent need for basic knowledge of ovarian tumor biology for the development of innovative EOC treatments.

Unlike melanoma or renal and hematologic tumor diseases, EOC is not considered to be immunogenic. However, there is evidence of an immune response against EOC in patients 4 . Experimental data show that the inflammatory microenvironment of EOC prevents the maturation of myeloid cells, favors regulatory cell development and restrains the cytotoxic activity of effector lymphocytes, leading the tumor to escape from the immune system and triggering cancer progression 5 . Treatments such as chemotherapy with paclitaxel/carboplatin and debulking surgery are traditionally considered to negatively impact the immune system during EOC 6 . However, recent data challenge this concept and highlight the major role of immune response in EOC. Indeed, aforementioned treatments were shown to modulate the host response and to decrease the immunosuppression 7 8 . Thus, immunotherapies aimed at increasing the host immune response or decreasing immunosuppression were tested in preclinical and clinical studies and are emerging as potential strategies to enhance classical EOC treatments.

In this article, we present an overview of the current understanding of the immune response and immune suppression in EOC. The immunological role of chemotherapy and surgery is highlighted, and pilot data supporting the efficiency of immunotherapy in EOC treatment are reviewed.

Evidence of an immune response in EOC

EOC expresses or overexpresses tumor-associated antigens (TAA), i.e. antigens (Ag) acquired by tumor cells in the process of neoplastic transformation that can elicit a specific T-cell immune response by the host. In 1993, EOC ascites were found to contain CD8+ T-cells capable of recognizing HER2/neu-positive tumor cells 9 . 5 to 66% of EOC exhibit this EGFR-related glycoprotein that activates signaling pathways involved in cellular proliferation 10 11 . Many other TAA were described in EOC, such as folate receptor(FR)-α 12 , epithelial cell adhesion molecule (EpCAM) 13 , human epididymis protein 4 14 , p53 15 , mucin-like MUC16 (CA125) and MUC1 (CA15.3) 16 and TAA of the cancer-testis group 17 18 . Tumor-reactive T-cells and antibodies (Ab) directed against TAA were detected in the peripheral blood of patients with advanced-stage disease at the time of diagnosis 15 19 , and tumor-reactive T-cells were isolated from tumors or ascites 20 .

Furthermore, there is clinical evidence for the role of immunosurveillance against EOC. The detection of intraepithelial tumor-infiltrating lymphocytes (TIL) correlates with clinical outcome. Zhang et al. detected CD3+ TIL in 102/186 frozen specimens from patients with stage III/IV EOC 21 . The five-year progression-free survival rates were 31.0% and 8.7% for patients with and without TIL, respectively. The presence of TIL correlated with progression-free survival in multivariate analysis (p < 0.001) 21 . Recently, other studies confirmed that the CD3+ TIL count is a significant prognosis factor in EOC (Table 1) 22 23 24 25 26 27 28 29 30 31 32 . High frequencies and activity levels of immune effector cells such as CD8+ T-cells, Natural Killer(NK)-cells and Vγ9Vδ2T-cells are correlated with positive clinical outcomes for EOC patients 33 34 . Thelper(Th)-17 cells, a recently discovered T-lymphocyte subset, were found in proportionally higher number in EOC microenvironment in comparison with other immune cells 35 36 . In EOC patients, Th17 levels in the tumor correlated positively with Th1-cells, cytotoxic CD8+ T-cells and NK-cells and Th17 levels in ascites correlated positively with patient survival 35 . Intriguingly, Th17 were reported to promote either tumor cell growth or antitumor response and their role in cancer development is currently under debate 37 . Finally, in addition to TIL, the number of peripheral blood immune cells, e.g. NK-cells, is also correlated with survival in EOC 33 . All these results support the existence of immunosurveillance in EOC (Figure 1).

<p>Table 1</p>

Authors

Year

Findings

PD-1: programmed cell death 1; PD-L1: PD-1 ligand 1; ETBR: endothelin B receptor; pDC: plasmacytoïd dendritic cells.

Spontaneous anti-tumor response

Zhang L et al. 21

2003

Association between intraepithelial T-cell infiltration (TIL CD3+) and patient survival

Raspollini NR et al. 22

2005

Association between intraepithelial T-cell infiltration (TIL CD3+) and patient survival (plus chemotherapeutic response)

Sato E et al. 23

2005

Association between intraepithelial T-cell infiltration (TIL CD8+) and patient survival

Hamanishi J et al. 24

2007

Association between intraepithelial T-cell infiltration (TIL CD3+) and patient survival

Clarke B et al. 25

2008

Association between intraepithelial T-cell infiltration (TIL CD8+) and patient survival (only for high grade serous EOC, but not for endometrioïd or mucinous EOC)

Shah CA et al. 26

2008

Association between intraepithelial T-cell infiltration (TIL CD8+) and optimal debulking surgery

Tomsova M et al. 27

2008

Association between intraepithelial T-cell infiltration (TIL CD3+) and patient survival

Callahan MJ et al. 38

2008

Association between intraepithelial T-cell infiltration (TIL CD8+) and patient survival

Han LY et al. 28

2008

Association between intraepithelial T-cell infiltration (TIL CD3+ and CD8+) and patient survival

Stumpf M et al. 29

2009

Association between intraepithelial T-cell infiltration (TIL CD3+ and CD8+) and patient survival

Leffers N et al. 30 39

2009

Association between intraepithelial T-cell infiltration (TIL CD8+) and patient survival

Milne K et al. 31

2009

Association between intraepithelial T-cell infiltration (TIL CD3+ and CD8+) and patient survival

Adams SF et al. 32

2009

Association between intraepithelial T-cell infiltration (TIL CD3+ and CD8+) and patient survival

Kryczek I et al. 35

2009

Association between intraepithelial T-cell infiltration (TIL CD4+ with IL-17 secretion) and patient survival

Tumor immune evasion

Curiel TJ et al. 36

2004

Inverse association between survival and intratumoral regulatory T cells (CD4+CD25+FoxP3+)

Wolf D et al. 40

2005

Inverse association between survival and intratumoral regulatory T cells (FoxP3+)

Dong HP et al. 41

2006

Inverse association between survival and intratumoral NK (CD3- CD16+) or B cells (CD19+)

Kryczek I et al. 42

2007

Inverse association between survival and intratumoral B7-H4+ macrophage or regulatory T cells (FoxP3+)

Hamanishi J et al. 24

2007

PD-L1 expression by tumor predicts low T-cell infiltration

Buckanovitch RJ et al. 43

2008

Endothelin B receptor (ETBR) expression restricts T-cell infiltration and predicts poor survival

Labidi-Galy SI 44 45

2011

Inverse association between survival and intratumoral pDC (CD4+, CD123+, BDCA2+)

Clinical arguments for the immunoediting hypothesis in epithelial ovarian carcinoma

<p>Figure 1</p>

Immune network in EOC

Immune network in EOC. EOC is immunogenic and expresses tumor-associated antigens such as HER2/neu, CA125 and Folate Receptor α. Various immune effectors such as CD8+ T-cells, NK-cells and Vγ9Vδ2T-cells can attack tumor cells, but immunosuppressive crosstalk counteracts the functions of these effector cells. Treg, tolerogenic DC, MDSC, B7-H4+ TAM and non-immune cells such as mesenchymal stem cells and tumor cells themselves halt antitumor activities through cell-cell contacts (CA125/siglec pathway, PD-1 and CTLA-4 immunosuppressive checkpoints) or the production of soluble factors (IL-10, TGF-β, PGE2, MIF, arginase-1, and IDO).

Immune escape in EOC

The tumor immunosurveillance concept was postulated in the 1960s by Burnet and Thomas, who proposed that the immune system patrols the body to recognize and destroy host cells that become cancerous and that the immune system is responsible for preventing cancer development 46 . This concept was then replaced by the cancer-immunoediting hypothesis, in which the immune system shapes tumor immunogenicity with three successive phases: elimination, equilibrium and escape 47 .

Immune escape in EOC involves several mechanisms that implicate tumor, immune and stromal cells. Ovarian tumor cells escape immune recognition by downregulating surface molecules involved in Ag presentation, such as β2-microglobulin and Major Histocompatibility Complex (MHC) 28 . Similarly, the downregulation of MHC class I-related chain A (MICA) expression impedes the detection of tumor cells by innate cytotoxic effector cells through the engagement of the NKG2D-activating receptor 48 49 . Additionally, ovarian tumor cells overexpress molecules that counteract the cytotoxic activities of immune cells: CA125 binds the NK-cell inhibitory receptor (KIR) siglec-9, thereby protecting themselves from NK-mediated lysis 50 51 ; the macrophage migration inhibitory factor (MIF) downregulates NKG2D-activating receptor expression on NK-cells 52 . Furthermore, engagement of programmed death-1 (PD-1) on CD8+ T-cells by programmed death-1 ligand-1 (PD-L1) expressed by ovarian tumor cells impairs the effector functions of these lymphocytes 24 53 . Wide panel of cancers, including EOC, were also shown to express indolamine-2,3-dioxygenase (IDO), an intracellular enzyme that catalyzes the rate-limiting step in the metabolism of the essential amino acid tryptophan 54 55 . IDO is a beneficial host mechanism regulating immune responses in various contexts such as pregnancy, transplantation or infection. It was proposed to elicit feedback process, therefore preventing deleterious consequences of excessive immune responses. However, this endogenous mechanism is hijacked by tumors to establish immunotolerance to tumor antigens 56 57 .

Immune cells also play a major role in the immune escape in EOC 58 . The EOC-specific recruitment of CD4+CD25+FoxP3+ regulatory T-cells (Treg), tolerogenic dendritic cells (DC), B7-H4+ tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) fosters immune privilege and predicts reduced survival in EOC (Table 1) 23 36 44 59 60 61 62 . Accumulation of Treg is now well documented in various tumors including EOC 23 36 38 39 40 41 . CCR4 chemokine receptor expression confers to Treg higher capacity than effector T-cells to infiltrate the tumor in response to CCL22 chemokine produced by either tumor cells or TAM 36 . In addition, Treg could originate from in situ expansion. In that setting, ICOS-ligand costimulation provided by plasmacytoid DC (pDC) was recently highlighted as a prominent signal triggering in situ Treg expansion in some tumors, including EOC 63 64 . At last, de novo conversion of FoxP3- cells into Treg was shown to occur in the tumor as a consequence of TGF-β stimulation or IDO induction 65 66 . Treg mainly mediate immunosuppression through cell-cell contacts with DC or effector cells or by the secretion of immunosuppressive cytokines, including IL-10, IL-35 and TGF-β 67 . Treg notably contribute to DC tolerization, thereby further reducing the effector T-cell activation and proliferation. Interestingly, association of tumor regulatory T-cells with high hazard ratio for death and decreased survival times is currently well documented in EOC 23 36 42 . Besides Treg, DC are instrumental in establishing immunosupression in cancer. While DC were initially recognized as the primary orchestrators of the immune response, their role in the immunotolerance is now well established 68 . Importantly, both conventional myeloid DC (cDC) and pDC are characterized by high plasticity 69 . Consequently, their immune properties could be modulated by environmental stimuli and tumors may benefit from this Achille’s heel to induce DC tolerization and to reduce the adaptive immunity to tumor antigens. Accordingly, studies showed that the EOC microenvironment converts DC toward an immunosuppressive phenotype 70 . In a mouse model of EOC, Scarlett et al. showed that the DC phenotype controls EOC progression. Indeed, the switch of infiltrating-DC from activating to regulatory phenotype coincides with rapid tumor progression to terminal disease 62 . The role of pDC in EOC immunity was proposed by Zou et al. that evidenced the recruitment of pDC in response to stromal-derived factor-1 (SDF-1/CXCL-12) secretion by EOC 71 . The accumulation of pDC within the EOC was shown to be associated with shorter progression-free survival 44 . Tolerogenic DC may exert profound immunosuppressive effects on effector lymphocytes. Alteration of the IFN-α production by pDC was recently documented in EOC 44 . Moreover, through PD-L1/PD-L2 expression, DC can engage the PD-1 inhibitory pathway, thus inhibiting lymphocyte proliferation and effector functions 72 73 , inducing tumor-specific T-cell apoptosis 74 and promoting the differentiation of CD4+ T-cells into Treg 75 . Tolerogenic DC can also turn-down the immune response through the induction of IDO activity that inhibits CD8+ T-cell proliferation 76 and decreases NKG2D expression on NK-cells 77 . As aforementioned for DC, the tumor microenvironment also strongly polarizes the macrophage differentiation and gives rise to TAM 37 . B7-H4+ macrophages, a subset of TAM, was shown to suppress TAA-specific T-cell immunity 60 . An inverse correlation was evidenced between the intensity of B7-H4 expression on macrophages in EOC and patient survival 42 . Moreover, average 5-year survival rate was found significantly higher in EOC patients with low densities of TAM than in patients with increased TAM populations 78 . At last, MDSC are immature myeloid cells with immunosuppressive properties that were evidenced in both mouse model of EOC and EOC patients 61 79 80 . MDSC exhibit increased level of arginase-1 (ARG-1) and inductible Nitric Oxide Synthase (iNOS) activities. Deprivation of L-Arginine in the tumor microenvironment is emerging as a key immunosuppressive mechanism. It leads to CD3-zeta chain downregulation, thereby inhibiting effector T-cell activation 81 . Increased levels of NO, along with reactive oxygen and nitrogen species, disrupt signaling through the IL-2 receptor 82 and alter Ag recognition by nitrating the TCR 83 . Moreover, MDSC were shown to facilitate effector T-cell conversion into Treg 84 and to inhibit intratumoral migration of CD8+ effectors because of the nitration of CCL2 chemoattractant 85 .

Third player in tumor escape is the stromal cell population. Overexpression of the endothelin-B receptor by tumor endothelial cells inhibits concurrent ICAM-1 expression, thereby impairing the ICAM-1/LFA-1-mediated transmigration of leukocytes 86 . Overexpression of the endothelin-B receptor is associated with the absence of TIL and short survival time in EOC patients 43 . Furthermore, stromal cells may provide chemoattractants for the immune cells e.g. SDF-1/CXCL12 that recruits pDC 71 . They are also able to secrete soluble immunosuppressive factors e.g. prostaglandin-E2 (PGE2) which is produced by mesenchymal stem cells (MSC).

Finally, the EOC microenvironment is characterized by the presence of numerous immunosuppressive soluble or cellular factors (IL-10, TGF-β, PGE2, MIF, HLA-G, IDO, arginase-1, PD-L1, B7-H4 and Fas-ligand), which can originate from various sources, including tumor, immune and stromal cells 87 88 89 90 91 . PGE2 can be secreted by both MSC and EOC tumor cells. Of note, overexpression of COX-2, an inducible enzyme that triggers PGE2 synthesis, by ovarian tumor cells correlates with resistance to chemotherapy and poor prognosis 92 . PGE2 inhibits NK and γδ T-cell cytotoxicity 45 93 94 and induces the differentiation of CD4+ T-cells into Treg 95 . Similarly, IDO is expressed in ovarian tumor cells and tumor-infiltrating DC 54 55 96 . IDO expression was reported in 43% of analyzed EOC tissues (83/192) 97 . Moreover, its expression was correlated with worse patient survival 54 55 and with enhanced peritoneal tumor dissemination 55 98 . IDO is currently thought as one of the main factors that contribute to tumor-induced immunosuppression by depleting tryptophan from the microenvironment and producing tryptophan metabolite kynurenine. Depletion of tryptophan is sensed by GCN2 kinase pathway driving effector T-cell anergy and apoptosis 99 . Effects of kynurenine are mediated by the aryl hydrocarbon receptor transcription factor that induces increased survival and motility in cancer cells while favoring Treg expansion and suppressive effects in effector T-cells 100 101 .

Thus, regulatory cells, along with soluble and cellular immunosuppressive factors, create a tolerogenic microenvironment in EOC that compromises the antitumoral immune response 89 . These EOC immunosuppressive networks characterize the “cancer immunoediting” concept, which emphasizes a dynamic process of interaction between cancer and the host immune system 47 (Figure 1).

Modulation of the immune response against EOC with debulking surgery or chemotherapy

Conventional EOC treatment uses debulking surgery and systemic chemotherapy. Surgery decreases the tumor burden and removes poorly vascularized tissues while cytotoxic drugs eradicate residual tumor cells 7 102 103 . Little information is available regarding the impact of surgery on the immunological status in EOC patients. Major surgery would induce immunosuppression because of the downregulation of T helper(Th)-1 response 6 104 . However, there is some evidence that tumor debulking reduces tumor-induced immunosuppression in EOC 7 105 . Napoletano et al. demonstrated that surgery significantly decreases the proportions of Treg and naive CD4+ T-cells while significantly increasing the ratio of CD8+ T-cells/Treg and the proportions of effector T-cells among the peripheral blood mononuclear cells 7 . Moreover, surgery significantly increases IFN-γ secretion by peripheral CD8+ T-cells and reduces the IL-10 immunosuppressive factor concentration in the serum 7 . Thus, cancer immunosuppression is partially reversible, and acquired immunity is enhanced by tumor debulking surgery in EOC 7 .

Regarding chemotherapy, the frequent induction of lymphopenia suggests that this treatment may be immunosuppressive. However, recent data indicate that immunity plays a major role in the therapeutic mechanisms associated with chemotherapy 106 107 . Accordingly, in advanced-EOC patients treated with platinum-based chemotherapy, an optimal tumor debulking outcome was more frequent when CD3+ TIL are present 21 . In addition, paclitaxel or cisplatin used in EOC cause the upregulation of mannose-6-phosphate receptor expression on murine tumor cells. This upregulation sensitizes tumor cells to granzyme-B protease released by cytotoxic T-lymphocytes 108 . Paclitaxel can also stimulate the proliferation of T-cells and enhance the cytolytic activity of NK-cells in models of breast cancer 106 109 . Moreover, in advanced EOC, successful chemotherapy was shown to be associated with improved functions and increased proportions of CD8+ effector T-cells 7 8 . Furthermore, chemotherapy decreases immunosuppression by reducing the number of circulating Treg observed after neoadjuvant chemotherapy in EOC 7 . Some antitumor agents can also trigger immunogenic tumor cell death, causing the cancer cells to expose or secrete immunogenic signals that trigger an anticancer immune response. Of note, not all types of chemotherapy, but oxaliplatin and 3/25 tested anthracyclines, elicit immunogenic cell death 110 111 112 . Altogether, these data provide evidence that debulking surgery and chemotherapy may restore, by direct and indirect effects, the equilibrium phase or the elimination phase in tumors that escaped initial immunosurveillance 106 .

Immunotherapy in EOC: how to counteract immunosuppression?

Preclinical and preliminary clinical studies aimed at proving the immunotherapy concept in EOC were initiated by using monoclonal Ab (mAb), vaccinations or adoptive T-cell transferts 113 114 115 . The majority of these studies were uncontrolled phase I/II studies, with small sample sizes and heterogeneous inclusion criteria (recurrent or chemotherapy-refractory diseases) disrupting the comparisons and the identification of the best strategy.

Several mAb targeting TAA were tested in EOC 116 : anti-CA125 oregovomab and abagovomab 117 118 119 ; anti-HER-2/neu trastuzumab and pertuzumab 10 120 ; anti-FR-α farletuzumab α 121 , anti-EpCAM catumaxomab 122 and anti-Tag72 B72.3 123 . All these mAb demonstrated adequate safety and tolerability but failed to demonstrate clear clinical benefits, even when an immunological response was evidenced 114 115 . Active immunotherapy by vaccination based on peptides or cellular approaches were also evaluated. Clinically tested peptides include NY-ESO-1 124 125 , p53 126 , HER2-neu 127 and multiple constructed-peptides (HER2-neu/MAGE-A1/FRα 128 and MUC-1/carcinoembryonic antigen 129 ). In addition, cell vaccines include DC pulsed with ARNm (FRα 130 ), peptides (HER2-neu, MUC1 131 ), autologous tumor Ag 132 or whole tumor cell lysate 133 . Vaccine therapies were well tolerated and demonstrated immunological responses, but provided only minor clinical benefits. Of note, these studies enrolled low numbers of patients and have generally not yet evolved past phase I/II studies. A third immunotherapy strategy is adoptive T-cell therapy, which uses cytotoxic lymphocytes with natural or engineered reactivity against cancer cells. Cytotoxic lymphocytes are generated in vitro and then transferred back into the patient to elicit cytotoxic responses against the patient’s own tumor cells. Only five phase I/II EOC studies, which enrolled few patients (<20), are available 134 135 136 137 138 . They used either TIL or peripheral autologous T-cells and were well tolerated; unfortunately, only modest clinical benefits were demonstrated. Thus, to date, results of these trials are disappointing, regardless of the strategy 115 . However, these trials may suffer from some pitfalls. First, they often enrolled patients with recurrent or refractory-chemotherapy diseases, i.e. patients at terminal stages of the disease, with strong immunodepression. It is likely that enrollment of patients at earlier stages of disease could be more successful. Secondly, all these trials focused on the recognition and killing of tumor cells and neglected to consider the immunosuppressive impact of the tumor microenvironment. Thus, improvement of theses immunotherapies is needed. For example, chimeric antigen receptor(CAR)-modified T cell therapy is highly promising 139 140 . CAR T-cells could be engineered to only express the downstream pathway of activating receptors. This refined adoptive therapy skips inhibitory signals expressed by the tumor environment. In addition, use of adjuvant drugs targeting immunosuppressive cells or soluble/cellular immunomodulatory factors could be the key to fully unleash the potential of immunotherapy by breaking peripheral tolerance.

Below, we review some immunomodulatory tools already in clinical use or likely to be assessed in the near future, that interact with the immunosuppressive factors found in the EOC microenvironment.

First approach may consist in depleting the host of the regulatory cells or in limiting their recruitment within the tumor. Treg depletion may be achieved using low-dose cyclophosphamide which prevents, under incompletely understood mechanism, Treg development and functionality 141 142 . An alternative strategy uses the expression by Treg of the IL-2 receptor alpha (CD25). Recombinant fusion protein of IL-2 and diphtheria toxin (Ontak®, Eisai) was tested in EOC patients and showed effective depletion of circulating Treg 143 . Moreover, in patients with metastatic breast cancer, the anti-CD25 mAb daclizumab (Zenapax®, Roche) demonstrated selective T-lymphocyte killing properties, allowing Treg depletion for several weeks 144 . However, it is unclear if Treg depletion occurs at EOC locations (solid tumor, malignant ascites) and results in tumor regression 143 144 145 . Moreover, as effector cells also express CD25, anti-CD25 mAb may also induce unwanted depletion of effector cells 146 . In addition, blocking the ICOS-pathway could inhibit the pDC-triggered proliferation of Treg within the tumor 64 . However, as ICOS pathway also favors the differentiation of T helper(Th)-17 cells which might either promote tumor growth or antitumor response 35 37 147 148 149 150 careful preclinical investigations of ICOS inhibitors (314.8 mAb) is needed 63 .

The role of chemoattractants in the recruitment of immune cells also gives a great opportunity to reduce the infiltration of regulatory cells within the tumor 151 . First candidates are under investigation. CCR4 antagonists were shown to block the recruitment of Treg instructed by CCL22 and CCL17 and to favor the induction of antigen-specific CD8+ T cell response after vaccination 152 . Similarly, Bindarit® that inhibits CCL2 synthesis and therefore restricts the recruitment in the tumor of immature myeloid cells, was shown to induce tumor regression in prostate and breast cancer animal models 153 . Regulatory cell depletion could also be achieved by improving the maturation of immature myeloid cells 154 using all trans retinoic acid 155 or ultra-low non-cytotoxic doses of paclitaxel (chemo-immunomodulation) 156 .

Another attractive approach is the use of either antagonists of immune-repressor molecules or agonists of immune-activating receptors 157 . Checkpoint blockade receptors comprise CTLA-4, PD-1 and NK inhibitory receptors (KIR) that, upon engagement, dampen the immune response. CTLA-4 predominantly regulates T-cells at the priming phase of activation by competing with CD28+ for binding of B7-1 and B7-2 on DC. CTLA-4 engagement prevents T-cells from achieving full activation. Accordingly, anti-CTLA-4 mAb were shown to activate CD4+ and CD8+ effector T-cells both directly by removing inhibitory checkpoints and indirectly via the inhibition of regulatory T-cell activity 158 . Eleven EOC patients, previously vaccinated with GM-CSF and irradiated autologous tumor cells, received anti-CTLA-4 ipilimumab (Yervoy®, Bristol-Myers-Squibb, BMS). Significant antitumor effects were observed in a minority of these patients and were correlated with increased CD8+ T-cells/Treg ratio 159 . In contrast to CTLA-4, PD-1 signaling occurs in the tumor, where PD-L1-expressing tumor cells can signal through PD-1 on TIL to turn-down the antitumor T-cell response. In EOC, the PD-1/PD-L1 pathway seems to be a dominant immunosuppression mechanism 73 . PD-L1 expression in EOC was demonstrated to be an independent unfavorable prognostic factor and to promote peritoneal dissemination 24 160 . Several PD-1/PD-L1-pathway blocking agents were assessed in various cancer types and promising results were recently reported. Nivolumab (BMS-936558, Bristol-Myers-Squibb) was tested in 296 patients most harboring lung cancer, renal cell cancer and melanoma, with clinical benefits apparent in 20 to 25% of the patients 161 . Impressive durable responses were reported. 25/42 patients with PD-L1-positive tumor experienced an objective response while none of the 17/42 PD-L1-negative patients did. However, lack of prognostic association was reported elsewhere and the usefulness of PD-L1 as a biomarker need to be explored in larger prospective studies 162 . In addition, Bramher and colleagues reported that 1/17 EOC patients treated with anti-PD-L1 mAb (BMS-936559) experienced an objective response 163 . New trials enrolling patients with solid tumor of multiple origins are underway and informative data in EOC are expected 164 . Inhibition of the cytotoxic properties of NK-cells through KIR engagement may also contribute to the tumor escape. Some anti-KIR antibodies, such as lilirumab (Bristol-Myers-Squibb), recently entered clinical development phases. First data were obtained in hematological diseases and phase I studies recruiting patients with solid tumors are ongoing 165 166 . As a corollary, agonistic agents of costimulatory molecules such as glucocorticoid-induced TNFR (GITR), OX40, CD137 are candidates to boost the antitumor immune response. A dose-escalation phase I clinical trial (NCT01239134) with agonist anti-GITR mAb (TRX518) was recently initiated.

Third possibility is to repress the activity of enzymes (IDO, ARG-1, iNOS) that were shown to inhibit the immune response. Data from first clinical trials using IDO inhibitors, notably the isomers of 1-methyl-tryptophan (1MT), were disappointing, but these studies may suffer from lack of potent and selective IDO inhibitors. New compounds recently entered clinical trials 167 . A phase II study of IDO inhibitor INCB024360 is currently recruiting patients with biochemical-recurrent-only EOC following complete remission with first-line chemotherapy (clinical trial: NCT01685255). In addition, inhibitors of phosphodiesterase(PDE)-5, e.g. sildenafil, were reported to increase intracellular concentrations of cGPM, resulting in the inhibition of both ARG-1 and iNOS. PDE-5 inhibitors along with nitroaspirin or specific ARG-1/iNOS inhibitors might provide new therapeutic strategy to recover potent antitumor immune response 154 .

Lastly, PGE2 was shown to be a crucial immunosuppressive factor in EOC, as it impairs the cytotoxic properties of effector cells such as Vγ9Vδ2T-cells 45 and also induces the differentiation of MDSC from bone marrow stem cells in a mouse model 168 . PGE2 biosynthesis is regulated by the inducible COX-2 enzyme and could be inhibited by the COX-2-specific inhibitor celecoxib (Celebrex®, Pfizer). In a mouse model, celecoxib prevented the local and systemic expansion of MDSC, impaired the suppressive function of these cells, and significantly improved vaccine immunotherapy 169 . Thus, celecoxib, currently used in the prevention of colorectal adenomatous polyps 170 , could be tested in combination with immunotherapy to reduce the immunosuppression by MDSC in EOC. Another possible strategy to counteract the immunosuppressive influence of PGE2 on Vγ9Vδ2T cells could be to restore the cytotoxic properties of these cells with a zoledronate perfusion 45 . In addition, zoledronate was shown to prevent the immunosuppressive polarization of TAM 171 172 which is a major component of the leukocyte infiltrate in the tumor microenvironment and plays a dominant role in the production of immune suppressive cytokines in EOC 60 . Thus, zoledronate, which is currently used for the management of osteoporosis and bone metastasis, appears to be an attractive molecule to reinforce the immune response. Altogether, these data warrant further exploration of combinatorial therapies with immunotherapy and bisphosphonates.

In conclusion, accumulated evidences support the immunoediting hypothesis and the idea that EOC is immunogenic. Immunotherapeutic protocols aimed at modulating the immune system to strengthen the spontaneous antitumor immune response are under investigation. Targeting the immunosuppressive mechanisms could be the key to fully unleash the potential of immunotherapy. The combination of molecules endowed with immuno-modulatory properties with immunotherapy targeting the tumor cells will hopefully increase the survival of EOC patients. Careful preclinical evaluation will be necessary to screen optimal combinations before clinical trials.

Competing interests

The authors declare that they have no competing interests.

Author’ contributions

VL, AT, FF, SH, VJ and FC reviewed the literature; VL, AT and FC wrote the paper; VL, JL, VC, MABR and FC proofread the final copy. All authors read and approved the final manuscript.

Acknowledgements

The authors thank American Journal Experts for editing the manuscript.

<p>Cancer statistics, 2009</p>JemalASiegelRWardEHaoYXuJThunMJCA Cancer J Clin20095922524910.3322/caac.2000619474385<p>Management of platinum-sensitive recurrent ovarian cancer</p>PfistererJLedermannJASemin Oncol200633S12S1616716798<p>Advanced ovarian cancer: phase III randomized study of sequential cisplatin-topotecan and carboplatin-paclitaxel vs carboplatin-paclitaxel</p>HoskinsPVergoteICervantesATuDStuartGZolaPPovedaAProvencherDKatsarosDOjedaBJ Natl Cancer Inst20101021547155610.1093/jnci/djq36220937992<p>Immunologic principles and immunotherapeutic approaches in ovarian cancer</p>KnutsonKLCurielTJSalazarLDisisMLHematol Oncol Clin North Am2003171051107310.1016/S0889-8588(03)00064-912959191<p>Blocking ovarian cancer progression by targeting tumor microenvironmental leukocytes</p>Cubillos-RuizJRRutkowskiMConejo-GarciaJRCell Cycle2010926026810.4161/cc.9.2.10430305620920023378<p>Downregulation of T helper type 1 immune response and altered pro-inflammatory and anti-inflammatory T cell cytokine balance following conventional but not laparoscopic surgery</p>BruneIBWilkeWHenslerTHolzmannBSiewertJRAm J Surg1999177556010.1016/S0002-9610(98)00299-210037309<p>Ovarian cancer cytoreduction induces changes in T cell population subsets reducing immunosuppression</p>NapoletanoCBellatiFLandiRPauselliSMarchettiCViscontiVSalePLiberatiMRughettiAFratiLJ Cell Mol Med2010142748275910.1111/j.1582-4934.2009.00911.x19780872<p>Recovery of CD8+ T-cell function during systemic chemotherapy in advanced ovarian cancer</p>ColemanSClaytonAMasonMDJasaniBAdamsMTabiZCancer Res2005657000700610.1158/0008-5472.CAN-04-379216061686<p>Cytotoxic T cells isolated from ovarian malignant ascites recognize a peptide derived from the HER-2/neu proto-oncogene</p>IoannidesCGFiskBFanDBiddisonWEWhartonJTO'BrianCACell Immunol199315122523410.1006/cimm.1993.12337691418<p>Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group</p>BookmanMADarcyKMClarke-PearsonDBoothbyRAHorowitzIRJ Clin Oncol20032128329010.1200/JCO.2003.10.10412525520<p>HER-2 overexpression is an independent marker of poor prognosis of advanced primary ovarian carcinoma: a multicenter study of the GINECO group</p>Camilleri-BroetSHardy-BessardACLe TourneauAParaisoDLevrelOLeducBBainSOrfeuvreHAudouinJPujade-LauraineEAnn Oncol20041510411210.1093/annonc/mdh02114679128<p>Ovarian cancer-associated lymphocyte recognition of folate binding protein peptides</p>PeoplesGEAndersonBWFiskBKudelkaAPWhartonJTIoannidesCGAnn Surg Oncol1998574375010.1007/BF023034869869522<p>Malignant ascites-derived exosomes of ovarian carcinoma patients contain CD24 and EpCAM</p>RunzSKellerSRuppCStoeckAIssaYKoensgenDMusteaASehouliJKristiansenGAltevogtPGynecol Oncol200710756357110.1016/j.ygyno.2007.08.06417900673<p>Human epididymis protein 4 (HE4) is a secreted glycoprotein that is overexpressed by serous and endometrioid ovarian carcinomas</p>DrapkinRvon HorstenHHLinYMokSCCrumCPWelchWRHechtJLCancer Res2005652162216910.1158/0008-5472.CAN-04-392415781627<p>Antibody immunity to the p53 oncogenic protein is a prognostic indicator in ovarian cancer</p>GoodellVSalazarLGUrbanNDrescherCWGrayHSwensenREMcIntoshMWDisisMLJ Clin Oncol20062476276810.1200/JCO.2005.03.281316391298<p>Aberrant expression of MUC4 in ovarian carcinoma: diagnostic significance alone and in combination with MUC1 and MUC16 (CA125)</p>ChauhanSCSinghAPRuizFJohanssonSLJainMSmithLMMoniauxNBatraSKMod Pathol2006191386139410.1038/modpathol.380064616880776<p>Expression of tumor-specific antigen MAGE, GAGE and BAGE in ovarian cancer tissues and cell lines</p>ZhangSZhouXYuHYuYBMC Cancer20101016310.1186/1471-2407-10-163286881120423514<p>Tumor vaccine for ovarian carcinoma targeting sperm protein 17</p>Chiriva-InternatiMWangZSalatiETimminsPLimSHCancer2002942447245310.1002/cncr.1050612015770<p>TRANCE- and CD40 ligand-matured dendritic cells reveal MHC class I-restricted T cells specific for autologous tumor in late-stage ovarian cancer patients</p>SchliengerKChuCSWooEYRiversPMTollAJHudsonBMausMVRileyJLChoiYCoukosGClin Cancer Res200391517152712684428<p>T-cell recognition of ovarian cancer</p>PeoplesGESchoofDDAndrewsJVGoedegebuurePSEberleinTJSurgery19931142272348342128<p>Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer</p>ZhangLConejo-GarciaJRKatsarosDGimottyPAMassobrioMRegnaniGMakrigiannakisAGrayHSchliengerKLiebmanMNN Engl J Med200334820321310.1056/NEJMoa02017712529460<p>Tumour-infiltrating gamma/delta T-lymphocytes are correlated with a brief disease-free interval in advanced ovarian serous carcinoma</p>RaspolliniMRCastiglioneFRossi Degl'innocentiDAmunniGVillanucciAGarbiniFBaroniGTaddeiGLAnn Oncol20051659059610.1093/annonc/mdi11215699022<p>Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer</p>SatoEOlsonSHAhnJBundyBNishikawaHQianFJungbluthAAFrosinaDGnjaticSAmbrosoneCProc Natl Acad Sci U S A2005102185381854310.1073/pnas.0509182102131174116344461<p>Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer</p>HamanishiJMandaiMIwasakiMOkazakiTTanakaYYamaguchiKHiguchiTYagiHTakakuraKMinatoNProc Natl Acad Sci U S A20071043360336510.1073/pnas.0611533104180558017360651<p>Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss</p>ClarkeBTinkerAVLeeCHSubramanianSvan de RijnMTurbinDKallogerSHanGCeballosKCadungogMGMod Pathol20092239340210.1038/modpathol.2008.19119060844<p>Intratumoral T cells, tumor-associated macrophages, and regulatory T cells: association with p53 mutations, circulating tumor DNA and survival in women with ovarian cancer</p>ShahCAAllisonKHGarciaRLGrayHJGoffBASwisherEMGynecol Oncol200810921521910.1016/j.ygyno.2008.01.01018314181<p>Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma</p>TomsovaMMelicharBSedlakovaISteinerIGynecol Oncol200810841542010.1016/j.ygyno.2007.10.01618037158<p>HLA class I antigen processing machinery component expression and intratumoral T-Cell infiltrate as independent prognostic markers in ovarian carcinoma</p>HanLYFletcherMSUrbauerDLMuellerPLandenCNKamatAALinYGMerrittWMSpannuthWADeaversMTClin Cancer Res2008143372337910.1158/1078-0432.CCR-07-4433342621618519766<p>Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: relevance of clonal selection of T lymphocytes</p>StumpfMHasenburgARienerMOJuttingUWangCShenYOrlowska-VolkMFischPWangZGitschGBr J Cancer20091011513152110.1038/sj.bjc.6605274277851719861998<p>Prognostic significance of tumor-infiltrating T-lymphocytes in primary and metastatic lesions of advanced stage ovarian cancer</p>LeffersNGoodenMJde JongRAHoogeboomBNten HoorKAHollemaHBoezenHMvan der ZeeAGDaemenTNijmanHWCancer Immunol Immunother20095844945910.1007/s00262-008-0583-518791714<p>Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors</p>MilneKKobelMKallogerSEBarnesROGaoDGilksCBWatsonPHNelsonBHPLoS One20094e641210.1371/journal.pone.0006412271276219641607<p>Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer</p>AdamsSFLevineDACadungogMGHammondRFacciabeneAOlveraNRubinSCBoydJGimottyPACoukosGCancer20091152891290210.1002/cncr.24317275481119472394<p>Natural killer cell activity and progression-free survival in ovarian cancer</p>GarzettiGGCignittiMCiavattiniAFabrisNRomaniniCGynecol Obstet Invest19933511812010.1159/0002926788383627<p>A quantitative deficiency in peripheral blood Vγ9Vδ2 cells is a negative prognostic biomarker in ovarian cancer patients</p>ThedrezALavouéVDessartheBDanielPHennoSJaffreILevêqueJCatrosVCabillicFPLoS One201385e63322Print 201310.1371/journal.pone.0063322366268823717410<p>Phenotype, distribution, generation, and functional and clinical relevance of Th17 cells in the human tumor environments</p>KryczekIBanerjeeMChengPVatanLSzeligaWWeiSHuangEFinlaysonESimeoneDWellingTHBlood20091141141114910.1182/blood-2009-03-208249272301119470694<p>Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival</p>CurielTJCoukosGZouLAlvarezXChengPMottramPEvdemon-HoganMConejo-GarciaJRZhangLBurowMNat Med20041094294910.1038/nm109315322536<p>Antigen-presenting cell (APC) subsets in ovarian cancer</p>WilkeCMKryczekIZouWInt Rev Immunol20113012012610.3109/08830185.2011.56736221557638<p>Increased HLA-DMB expression in the tumor epithelium is associated with increased CTL infiltration and improved prognosis in advanced-stage serous ovarian cancer</p>CallahanMJNagymanyokiZBonomeTJohnsonMELitkouhiBSullivanEHHirschMSMatulonisUALiuJBirrerMJClin Cancer Res2008147667767310.1158/1078-0432.CCR-08-0479300016519047092<p>Identification of genes and pathways associated with cytotoxic T lymphocyte infiltration of serous ovarian cancer</p>LeffersNFehrmannRSGoodenMJSchulzeURTen HoorKAHollemaHBoezenHMDaemenTde JongSNijmanHWvan der ZeeAGBr J Cancer201010368569210.1038/sj.bjc.6605820293826220664601<p>The expression of the regulatory T cell-specific forkhead box transcription factor FoxP3 is associated with poor prognosis in ovarian cancer</p>WolfDWolfAMRumpoldHFieglHZeimetAGMuller-HolznerEDeiblMGastlGGunsiliusEMarthCClin Cancer Res2005118326833110.1158/1078-0432.CCR-05-124416322292<p>NK- and B-cell infiltration correlates with worse outcome in metastatic ovarian carcinoma</p>DongHPElstrandMBHolthASilinsIBernerATropeCGDavidsonBRisbergBAm J Clin Pathol200612545145816613351<p>Relationship between B7-H4, regulatory T cells, and patient outcome in human ovarian carcinoma</p>KryczekIWeiSZhuGMyersLMottramPChengPChenLCoukosGZouWCancer Res2007678900890510.1158/0008-5472.CAN-07-186617875732<p>Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy</p>BuckanovichRJFacciabeneAKimSBenenciaFSasaroliDBalintKKatsarosDO'Brien-JenkinsAGimottyPACoukosGNat Med200814283610.1038/nm169918157142<p>Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer</p>Labidi-GalySISisirakVMeeusPGobertMTreilleuxIBajardACombesJDFagetJMithieuxFCassignolACancer Res2011715423543410.1158/0008-5472.CAN-11-036721697280<p>Sensitization of ovarian carcinoma cells with zoledronate restores the cytotoxic capacity of Vγ9Vδ2 T cells impaired by the prostaglandin E2 immunosuppressive factor: implications for immunotherapy</p>LavouéVCabillicFToutiraisOThedrezADessartheBde La PintièreCTDanielPFoucherFBauvilleEHennoSBurtinFBansardJYLevêqueJCatrosVBouet-ToussaintFInt J Cancer20121314E449E462Epub 2011 Dec 2110.1002/ijc.2735322095289<p>The concept of immunological surveillance</p>BurnetFMProg Exp Tumor Res1970131274921480<p>Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion</p>SchreiberRDOldLJSmythMJScience20113311565157010.1126/science.120348621436444<p>Human ovarian tumor cells escape gammadelta T cell recognition partly by down regulating surface expression of MICA and limiting cell cycle related molecules</p>LuJAggarwalRKanjiSDasMJosephMPompiliVDasHPLoS One20116e2334810.1371/journal.pone.0023348317335621935360<p>Self/non-self discrimination by human gammadelta T cells: simple solutions for a complex issue?</p>ThedrezASabourinCGertnerJDevilderMCAllain-MailletSFournieJJScotetEBonnevilleMImmunol Rev200721512313510.1111/j.1600-065X.2006.00468.x17291284<p>MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells</p>GubbelsJAFelderMHoribataSBelisleJAKapurAHoldenHPetrieSMigneaultMRancourtCConnorJPPatankarMSMol Cancer201091110.1186/1476-4598-9-11281869320089172<p>Identification of Siglec-9 as the receptor for MUC16 on human NK cells, B cells, and monocytes</p>BelisleJAHoribataSJenniferGAPetrieSKapurAAndreSGabiusHJRancourtCConnorJPaulsonJCPatankarMSMol Cancer2010911810.1186/1476-4598-9-118289060420497550<p>Macrophage migration inhibitory factor contributes to the immune escape of ovarian cancer by down-regulating NKG2D</p>KrockenbergerMDombrowskiYWeidlerCOssadnikMHonigAHauslerSVoigtHBeckerJCLengLSteinleAJ Immunol200818073387348360774218490733<p>Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer</p>MatsuzakiJGnjaticSMhawech-FaucegliaPBeckAMillerATsujiTEppolitoCQianFLeleSShrikantPProc Natl Acad Sci U S A20101077875788010.1073/pnas.1003345107286790720385810<p>Indoleamine 2,3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells</p>OkamotoANikaidoTOchiaiKTakakuraSSaitoMAokiYIshiiNYanaiharaNYamadaKTakikawaOClin Cancer Res2005116030603910.1158/1078-0432.CCR-04-267116115948<p>Role of the immunosuppressive enzyme indoleamine 2,3-dioxygenase in the progression of ovarian carcinoma</p>InabaTInoKKajiyamaHYamamotoEShibataKNawaANagasakaTAkimotoHTakikawaOKikkawaFGynecol Oncol200911518519210.1016/j.ygyno.2009.07.01519665763<p>Creating immune privilege: active local suppression that benefits friends, but protects foes</p>MellorALMunnDHNat Rev Immunol20088748010.1038/nri223318064049<p>Host indoleamine 2,3-dioxygenase: contribution to systemic acquired tumor tolerance</p>JohnsonTSMunnDHImmunol Invest20124176579710.3109/08820139.2012.68940523017145<p>Cross-talk between myeloid-derived suppressor cells (MDSC), macrophages, and dendritic cells enhances tumor-induced immune suppression</p>Ostrand-RosenbergSSinhaPBeuryDWClementsVKSemin Cancer Biol20122227528110.1016/j.semcancer.2012.01.01122313874<p>Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity</p>CurielTJWeiSDongHAlvarezXChengPMottramPKrzysiekRKnutsonKLDanielBZimmermannMCNat Med2003956256710.1038/nm86312704383<p>B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma</p>KryczekIZouLRodriguezPZhuGWeiSMottramPBrumlikMChengPCurielTMyersLJ Exp Med200620387188110.1084/jem.20050930211830016606666<p>CD80 in immune suppression by mouse ovarian carcinoma-associated Gr-1 + CD11b + myeloid cells</p>YangRCaiZZhangYYutzyWHRobyKFRodenRBCancer Res2006666807681510.1158/0008-5472.CAN-05-375516818658<p>Ovarian cancer progression is controlled by phenotypic changes in dendritic cells</p>ScarlettUKRutkowskiMRRauwerdinkAMFieldsJEscovar-FadulXBairdJCubillos-RuizJRJacobsACGonzalezJLWeaverJJ Exp Med201220949550610.1084/jem.20111413330223422351930<p>ICOS-ligand expression on plasmacytoid dendritic cells supports breast cancer progression by promoting the accumulation of immunosuppressive CD4+ T cells</p>FagetJBendriss-VermareNGobertMDurandIOliveDBiotaCBachelotTTreilleuxIGoddard-LeonSLavergneECancer Res2012726130614110.1158/0008-5472.CAN-12-240923026134<p>Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells</p>ConradCGregorioJWangYHItoTMellerSHanabuchiSAndersonSAtkinsonNRamirezPTLiuYJCancer Res2012725240524910.1158/0008-5472.CAN-12-227122850422<p>Tumor evasion of the immune system by converting CD4 + CD25- T cells into CD4 + CD25+ T regulatory cells: role of tumor-derived TGF-beta</p>LiuVCWongLYJangTShahAHParkIYangXZhangQLonningSTeicherBALeeCJ Immunol20071782883289217312132<p>Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells</p>CurtiAPandolfiSValzasinaBAluigiMIsidoriAFerriESalvestriniVBonannoGRutellaSDurelliIBlood20071092871287717164341<p>Suppression, subversion and escape: the role of regulatory T cells in cancer progression</p>OleinikaKNibbsRJGrahamGJFraserARClin Exp Immunol2013171364510.1111/j.1365-2249.2012.04657.x23199321<p>Tolerogenic dendritic cells</p>SteinmanRMHawigerDNussenzweigMCAnnu Rev Immunol20032168571110.1146/annurev.immunol.21.120601.14104012615891<p>Functional diversity and plasticity of human dendritic cell subsets</p>ItoTLiuYJKadowakiNInt J Hematol20058118819610.1532/IJH97.0501215814329<p>In situ stimulation of CD40 and Toll-like receptor 3 transforms ovarian cancer-infiltrating dendritic cells from immunosuppressive to immunostimulatory cells</p>ScarlettUKCubillos-RuizJRNesbethYCMartinezDGEngleXGewirtzATAhonenCLConejo-GarciaJRCancer Res2009697329733710.1158/0008-5472.CAN-09-0835275480619738057<p>Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells</p>ZouWMachelonVCoulomb-L'HerminABorvakJNomeFIsaevaTWeiSKrzysiekRDurand-GasselinIGordonANat Med200171339134610.1038/nm1201-133911726975<p>B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells</p>TsengSYOtsujiMGorskiKHuangXSlanskyJEPaiSIShalabiAShinTPardollDMTsuchiyaHJ Exp Med200119383984610.1084/jem.193.7.839219337011283156<p>Tumor-infiltrating programmed death receptor-1+ dendritic cells mediate immune suppression in ovarian cancer</p>KrempskiJKaryampudiLBehrensMDErskineCLHartmannLDongHGoodeELKalliKRKnutsonKLJ Immunol20111866905691310.4049/jimmunol.1100274311054921551365<p>Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion</p>DongHStromeSESalomaoDRTamuraHHiranoFFliesDBRochePCLuJZhuGTamadaKNat Med2002879380012091876<p>Programmed death 1 ligand signaling regulates the generation of adaptive Foxp3 + CD4+ regulatory T cells</p>WangLPino-LagosKde VriesVCGuleriaISayeghMHNoelleRJProc Natl Acad Sci U S A20081059331933610.1073/pnas.0710441105244281718599457<p>Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes</p>MunnDHSharmaMDHouDBabanBLeeJRAntoniaSJMessinaJLChandlerPKoniPAMellorALJ Clin Invest200411428029044975015254595<p>The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function</p>Della ChiesaMCarlomagnoSFrumentoGBalsamoMCantoniCConteRMorettaLMorettaAVitaleMBlood20061084118412510.1182/blood-2006-03-00670016902152<p>[Prognostic significance of tumor-associated macrophage infiltration in advanced epithelial ovarian carcinoma]</p>WanTLiuJHZhengLMCaiMYDingTAi Zheng20092832332719619451<p>Murine ovarian cancer vascular leukocytes require arginase-1 activity for T cell suppression</p>BakSPAlonsoATurkMJBerwinBMol Immunol20084625826810.1016/j.molimm.2008.08.266261319318824264<p>PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment</p>ObermajerNMuthuswamyROdunsiKEdwardsRPKalinskiPCancer Res2011717463747010.1158/0008-5472.CAN-11-244922025564<p>TCR zeta down-regulation under chronic inflammation is mediated by myeloid suppressor cells differentially distributed between various lymphatic organs</p>EzernitchiAVVakninICohen-DanielLLevyOManasterEHalabiAPikarskyEShapiraLBaniyashMJ Immunol20061774763477216982917<p>Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism</p>MazzoniABronteVVisintinASpitzerJHApolloniESerafiniPZanovelloPSegalDMJ Immunol200216868969511777962<p>Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer</p>NagarajSGuptaKPisarevVKinarskyLShermanSKangLHerberDLSchneckJGabrilovichDINat Med20071382883510.1038/nm1609213560717603493<p>Gr-1 + CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host</p>HuangBPanPYLiQSatoAILevyDEBrombergJDivinoCMChenSHCancer Res2006661123113110.1158/0008-5472.CAN-05-129916424049<p>Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells</p>MolonBUgelSDel PozzoFSoldaniCZilioSAvellaDDe PalmaAMauriPMonegalARescignoMJ Exp Med20112081949196210.1084/jem.20101956318205121930770<p>ICAM-1 regulates neutrophil adhesion and transcellular migration of TNF-alpha-activated vascular endothelium under flow</p>YangLFroioRMSciutoTEDvorakAMAlonRLuscinskasFWBlood200510658459210.1182/blood-2004-12-4942163524115811956<p>Soluble HLA-G molecules are increased in lymphoproliferative disorders</p>SebtiYLe FriecGPangaultCGrosFDrenouBGuillouxVBernardMLamyTFauchetRAmiotLHum Immunol2003641093110110.1016/j.humimm.2003.08.34514602240<p>Peritoneal inflammation - A microenvironment for Epithelial Ovarian Cancer (EOC)</p>FreedmanRSDeaversMLiuJWangEJ Transl Med200422310.1186/1479-5876-2-2345952115219235<p>The impact of T-cell immunity on ovarian cancer outcomes</p>NelsonBHImmunol Rev200822210111610.1111/j.1600-065X.2008.00614.x18363996<p>Human leukocyte antigen-G expression is associated with a poor prognosis in patients with esophageal squamous cell carcinoma</p>LinAZhangXZhouWJRuanYYXuDPWangQYanWHInt J Cancer20111291382139010.1002/ijc.2580721128238<p>Regulation of invasion of epithelial ovarian cancer by transforming growth factor-beta</p>RodriguezGCHaisleyCHurteauJMoserTLWhitakerRBastRCJrStackMSGynecol Oncol20018024525310.1006/gyno.2000.604211161867<p>Increased cyclooxygenase-2 (COX-2) expression is associated with chemotherapy resistance and outcome in ovarian cancer patients</p>FerrandinaGLauriolaLZannoniGFFagottiAFanfaniFLeggeFMaggianoNGessiMMancusoSRanellettiFOScambiaGAnn Oncol2002131205121110.1093/annonc/mdf20712181243<p>PGE2 inhibits natural killer and gamma delta T cell cytotoxicity triggered by NKR and TCR through a cAMP-mediated PKA type I-dependent signaling</p>MartinetLJeanCDietrichGFournieJJPoupotRBiochem Pharmacol20108083884510.1016/j.bcp.2010.05.00220470757<p>Mesenchymal progenitor cells as cellular vehicles for delivery of oncolytic adenoviruses</p>KomarovaSKawakamiYStoff-KhaliliMACurielDTPereboevaLMol Cancer Ther2006575576610.1158/1535-7163.MCT-05-033416546991<p>Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells</p>BaratelliFLinYZhuLYangSCHeuze-Vourc'hNZengGReckampKDohadwalaMSharmaSDubinettSMJ Immunol20051751483149016034085<p>Indoleamine 2,3-dioxygenase and dendritic cell tolerogenicity</p>HardenJLEgilmezNKImmunol Invest20124173876410.3109/08820139.2012.676122364591223017144<p>Efficacy of levo-1-methyl tryptophan and dextro-1-methyl tryptophan in reversing indoleamine-2,3-dioxygenase-mediated arrest of T-cell proliferation in human epithelial ovarian cancer</p>QianFVillellaJWallacePKMhawech-FaucegliaPTarioJDJrAndrewsCMatsuzakiJValmoriDAyyoubMFrederickPJCancer Res2009695498550410.1158/0008-5472.CAN-08-210619491279<p>Indoleamine 2,3-dioxygenase promotes peritoneal dissemination of ovarian cancer through inhibition of natural killercell function and angiogenesis promotion</p>NonakaHSagaYFujiwaraHAkimotoHYamadaAKagawaSTakeiYMachidaSTakikawaOSuzukiMInt J Oncol20113811312021109932<p>GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase</p>MunnDHSharmaMDBabanBHardingHPZhangYRonDMellorALImmunity20052263364210.1016/j.immuni.2005.03.01315894280<p>The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells</p>FallarinoFGrohmannUYouSMcGrathBCCavenerDRVaccaCOrabonaCBianchiRBelladonnaMLVolpiCJ Immunol20061766752676116709834<p>Tryptophan catabolism in cancer: beyond IDO and tryptophan depletion</p>PlattenMWickWVan den EyndeBJCancer Res20117254355440<p>Tumor residual after surgical cytoreduction in prediction of clinical outcome in stage IV epithelial ovarian cancer: a Gynecologic Oncology Group Study</p>WinterWE3rdMaxwellGLTianCSundborgMJRoseGSRosePGRubinSCMuggiaFMcGuireWPJ Clin Oncol200826838910.1200/JCO.2007.13.195318025437<p>A critique of surgical cytoreduction in advanced ovarian cancer</p>CovensALGynecol Oncol20007826927410.1006/gyno.2000.592610985879<p>Distinct mechanisms of immunosuppression as a consequence of major surgery</p>HenslerTHeckerHHeegKHeideckeCDBartelsHBarthlenWWagnerHSiewertJRHolzmannBInfect Immun199765228322911753179169765<p>Suppressing the suppressor: Role of immunosuppressive regulatory T cells in cancer surgery</p>BaumgartnerJMMcCarterMDSurgery200914534535010.1016/j.surg.2008.12.01319303982<p>Immune parameters affecting the efficacy of chemotherapeutic regimens</p>ZitvogelLKeppOKroemerGNat Rev Clin Oncol2011815116010.1038/nrclinonc.2010.22321364688<p>Contribution of IL-17-producing gamma delta T cells to the efficacy of anticancer chemotherapy</p>MaYAymericLLocherCMattarolloSRDelahayeNFPereiraPBoucontetLApetohLGhiringhelliFCasaresNJ Exp Med201120849150310.1084/jem.20100269305857521383056<p>Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice</p>RamakrishnanRAssudaniDNagarajSHunterTChoHIAntoniaSAltiokSCelisEGabrilovichDIJ Clin Invest20101201111112410.1172/JCI40269284604820234093<p>Cellular immunity in breast cancer patients completing taxane treatment</p>CarsonWE3rdShapiroCLCrespinTRThorntonLMAndersenBLClin Cancer Res2004103401340910.1158/1078-0432.CCR-1016-0315161695<p>The anticancer immune response: indispensable for therapeutic success?</p>ZitvogelLApetohLGhiringhelliFAndreFTesniereAKroemerGJ Clin Invest20081181991200110.1172/JCI35180239690518523649<p>Immunogenic cancer cell death: a key-lock paradigm</p>TesniereAApetohLGhiringhelliFJozaNPanaretakisTKeppOSchlemmerFZitvogelLKroemerGCurr Opin Immunol20082050451110.1016/j.coi.2008.05.00718573340<p>Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death</p>CasaresNPequignotMOTesniereAGhiringhelliFRouxSChaputNSchmittEHamaiAHervas-StubbsSObeidMJ Exp Med20052021691170110.1084/jem.20050915221296816365148<p>Immunotherapy for ovarian cancer: what's next?</p>KandalaftLEPowellDJJrSinghNCoukosGJ Clin Oncol20112992593310.1200/JCO.2009.27.2369306806421079136<p>Immunity and immune suppression in human ovarian cancer</p>PrestonCCGoodeELHartmannLCKalliKRKnutsonKLImmunotherapy2011353955610.2217/imt.11.20314714421463194<p>Antigen-specific active immunotherapy for ovarian cancer</p>LeffersNDaemenTHelfrichWBoezenHMCohlenBJMeliefKNijmanHWCochrane Database Syst Rev20101CD007287Review. PMID: 20091627 [PubMed - indexed for MEDLINE]10.1002/14651858.CD007287.pub2<p>Preclinical studies and clinical utilization of monoclonal antibodies in epithelial ovarian cancer</p>FrederickPJStraughnJMJrAlvarezRDBuchsbaumDJGynecol Oncol200911338439010.1016/j.ygyno.2009.01.00819232697<p>Oregovomab maintenance monoimmunotherapy does not improve outcomes in advanced ovarian cancer</p>BerekJTaylorPMcGuireWSmithLMSchultesBNicodemusCFJ Clin Oncol20092741842519075271<p>Vaccination of patients with advanced ovarian carcinoma with the anti-idiotype ACA125: immunological response and survival (phase Ib/II)</p>ReinartzSKohlerSSchlebuschHKristaKGiffelsPRenkeKHuoberJMobusVKreienbergRDuBoisAClin Cancer Res2004101580158710.1158/1078-0432.CCR-03-005615014007<p>The anti-idiotypic antibody abagovomab in patients with recurrent ovarian cancer. A phase I trial of the AGO-OVAR</p>PfistererJdu BoisASehouliJLoiblSReinartzSReussACanzlerUBelauAJackischCKimmigRAnn Oncol2006171568157710.1093/annonc/mdl35717005631<p>Clinical activity of gemcitabine plus pertuzumab in platinum-resistant ovarian cancer, fallopian tube cancer, or primary peritoneal cancer</p>MakhijaSAmlerLCGlennDUelandFRGoldMADizonDSPatonVLinCYJanuarioTNgKJ Clin Oncol2010281215122310.1200/JCO.2009.22.335419901115<p>Farletuzumab, a humanized monoclonal antibody against folate receptor alpha, in epithelial ovarian cancer: a phase I study</p>KonnerJABell-McGuinnKMSabbatiniPHensleyMLTewWPPandit-TaskarNVander ElsNPhillipsMDSchweizerCWeilSCClin Cancer Res2010165288529510.1158/1078-0432.CCR-10-070020855460<p>The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: Results of a prospective randomized phase II/III trial</p>HeissMMMurawaPKoralewskiPKutarskaEKolesnikOOIvanchenkoVVDudnichenkoASAleknavicieneBRazbadauskasAGoreMInt J Cancer20101272209222110.1002/ijc.25423295845820473913<p>Phase I study of 90Y-labeled B72.3 intraperitoneal administration in patients with ovarian cancer: effect of dose and EDTA coadministration on pharmacokinetics and toxicity</p>RosenblumMGVerschraegenCFMurrayJLKudelkaAPGanoJCheungLKavanaghJJClin Cancer Res1999595396110353726<p>Safety and immunogenicity study of NY-ESO-1b peptide and montanide ISA-51 vaccination of patients with epithelial ovarian cancer in high-risk first remission</p>DiefenbachCSGnjaticSSabbatiniPAghajanianCHensleyMLSpriggsDRIasonosALeeHDupontBPezzulliSClin Cancer Res2008142740274810.1158/1078-0432.CCR-07-461918451240<p>Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer</p>OdunsiKQianFMatsuzakiJMhawech-FaucegliaPAndrewsCHoffmanEWPanLRitterGVillellaJThomasBProc Natl Acad Sci U S A2007104128371284210.1073/pnas.0703342104193755317652518<p>Immunization with a P53 synthetic long peptide vaccine induces P53-specific immune responses in ovarian cancer patients, a phase II trial</p>LeffersNLambeckAJGoodenMJHoogeboomBNWolfRHammingIEHepkemaBGWillemsePHMolmansBHHollemaHInt J Cancer20091252104211310.1002/ijc.2459719621448<p>Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines</p>DisisMLGooleyTARinnKDavisDPiepkornMCheeverMAKnutsonKLSchiffmanKJ Clin Oncol2002202624263210.1200/JCO.2002.06.17112039923<p>A multipeptide vaccine is safe and elicits T-cell responses in participants with advanced stage ovarian cancer</p>Chianese-BullockKAIrvinWPJrPetroniGRMurphyCSmolkinMOlsonWCColemanEBoernerSANailCJNeesePYJ Immunother20083142043010.1097/CJI.0b013e31816dad1018391753<p>Pilot study of vaccination with recombinant CEA-MUC-1-TRICOM poxviral-based vaccines in patients with metastatic carcinoma</p>GulleyJLArlenPMTsangKYYokokawaJPalenaCPooleDJRemondoCCeredaVJonesJLPazdurMPClin Cancer Res2008143060306910.1158/1078-0432.CCR-08-0126267309718483372<p>Vaccination with dendritic cells transfected with mRNA-encoded folate-receptor-alpha for relapsed metastatic ovarian cancer</p>HernandoJJParkTWFischerHPZivanovicOBraunMPolcherMGrunnULeutnerCPotzschBKuhnWLancet Oncol2007845145410.1016/S1470-2045(07)70142-017466904<p>Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells</p>BrossartPWirthsSStuhlerGReichardtVLKanzLBruggerWBlood2000963102310811049990<p>Vaccination with autologous tumour antigen-pulsed dendritic cells in advanced gynaecological malignancies: clinical and immunological evaluation of a phase I trial</p>HernandoJJParkTWKublerKOffergeldRSchlebuschHBauknechtTCancer Immunol Immunother200251455210.1007/s00262-001-0255-111845259<p>Induction of T cell responses against autologous ovarian tumors with whole tumor cell lysate-pulsed dendritic cells</p>ZhaoXWeiYQPengZLImmunol Invest200130334510.1081/IMM-10010368911419910<p>Use of adoptive transfer of tumor-infiltrating lymphocytes alone or in combination with cisplatin-containing chemotherapy in patients with epithelial ovarian cancer</p>AokiYTakakuwaKKodamaSTanakaKTakahashiMTokunagaATakahashiTCancer Res199151193419392004379<p>Intraperitoneal adoptive immunotherapy of ovarian carcinoma with tumor-infiltrating lymphocytes and low-dose recombinant interleukin-2: a pilot trial</p>FreedmanRSEdwardsCLKavanaghJJKudelkaAPKatzRLCarrascoCHAtkinsonENScottWTomasovicBTemplinSJ Immunother Emphasis Tumor Immunol19941619821010.1097/00002371-199410000-000047834119<p>Prolonged disease-free period in patients with advanced epithelial ovarian cancer after adoptive transfer of tumor-infiltrating lymphocytes</p>FujitaKIkarashiHTakakuwaKKodamaSTokunagaATakahashiTTanakaKClin Cancer Res199515015079816009<p>A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer</p>KershawMHWestwoodJAParkerLLWangGEshharZMavroukakisSAWhiteDEWunderlichJRCanevariSRogers-FreezerLClin Cancer Res2006126106611510.1158/1078-0432.CCR-06-1183215435117062687<p>Autologous MUC1-specific Th1 effector cell immunotherapy induces differential levels of systemic TReg cell subpopulations that result in increased ovarian cancer patient survival</p>DobrzanskiMJRewers-FelkinsKAQuinlinISSamadKAPhillipsCARobinsonWDobrzanskiDJWrightSEClin Immunol200913333335210.1016/j.clim.2009.08.007280508519762283<p>Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions</p>CurranKJPegramHJBrentjensRJJ Gene Med20121440541522262649<p>The basic principles of chimeric antigen receptor design</p>SadelainMBrentjensRRiviereICancer Discov2013338839810.1158/2159-8290.CD-12-054823550147<p>Induction of cell-mediated immunity to autologous melanoma cells and regression of metastases after treatment with a melanoma cell vaccine preceded by cyclophosphamide</p>BerdDMaguireHCJrMastrangeloMJCancer Res198646257225773697996<p>Metronomic cyclophosphamide regimen selectively depletes CD4 + CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients</p>GhiringhelliFMenardCPuigPELadoireSRouxSMartinFSolaryELe CesneAZitvogelLChauffertBCancer Immunol Immunother20075664164810.1007/s00262-006-0225-816960692<p>Inability of a fusion protein of IL-2 and diphtheria toxin (Denileukin Diftitox, DAB389IL-2, ONTAK) to eliminate regulatory T lymphocytes in patients with melanoma</p>AttiaPMakerAVHaworthLRRogers-FreezerLRosenbergSAJ Immunother20052858259210.1097/01.cji.0000175468.19742.10153376416224276<p>CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients</p>RechAJMickRMartinSRecioAAquiNAPowellDJJrColligonTATroskoJALeinbachLIPletcherCHSci Transl Med20124134ra162<p>Regulatory T cells in ovarian cancer: biology and therapeutic potential</p>BarnettBKryczekIChengPZouWCurielTJAm J Reprod Immunol20055436937710.1111/j.1600-0897.2005.00330.x16305662<p>Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses</p>SutmullerRPvan DuivenvoordeLMvan ElsasASchumacherTNWildenbergMEAllisonJPToesREOffringaRMeliefCJJ Exp Med200119482383210.1084/jem.194.6.823219595511560997<p>Th17 cells in cancer: help or hindrance?</p>WilkeCMKryczekIWeiSZhaoEWuKWangGZouWCarcinogenesis20113264364910.1093/carcin/bgr019308669921304053<p>Human Th17 cells in patients with cancer: Friends or foe?</p>GretenTFZhaoFGamrekelashviliJKorangyFOncoimmunology201211438143910.4161/onci.21245351853023243621<p>T helper 17 cells promote cytotoxic T cell activation in tumor immunity</p>Martin-OrozcoNMuranskiPChungYYangXOYamazakiTLuSHwuPRestifoNPOverwijkWWDongCImmunity20093178779810.1016/j.immuni.2009.09.014278778619879162<p>The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans</p>CharlesKAKulbeHSoperREscorcio-CorreiaMLawrenceTSchultheisAChakravartyPThompsonRGKolliasGSmythJFJ Clin Invest20091193011302310.1172/JCI39065275207619741298<p>Chemokine receptors as targets for cancer therapy</p>WuXLeeVCChevalierEHwangSTCurr Pharm Des20091574275710.2174/13816120978758216519275640<p>A CCR4 antagonist combined with vaccines induces antigen-specific CD8+ T cells and tumor immunity against self antigens</p>PereHMontierYBayryJQuintin-ColonnaFMerillonNDransartEBadoualCGeyARavelPMarcheteauEBlood20111184853486210.1182/blood-2011-01-32965621908423<p>Targeting monocyte chemotactic protein-1 synthesis with bindarit induces tumor regression in prostate and breast cancer animal models</p>ZolloMDi DatoVSpanoDDe MartinoDLiguoriLMarinoNVastoloVNavasLGarroneBManganoGClin Exp Metastasis20122958560110.1007/s10585-012-9473-522484917<p>Myeloid-derived suppressor cells interact with tumors in terms of myelopoiesis, tumorigenesis and immunosuppression: thick as thieves</p>SevkoAUmanskyVJ Cancer2013431110.7150/jca.5047356424223386900<p>All-trans-retinoic acid improves differentiation of myeloid cells and immune response in cancer patients</p>MirzaNFishmanMFrickeIDunnMNeugerAMFrostTJLushRMAntoniaSGabrilovichDICancer Res2006669299930710.1158/0008-5472.CAN-06-1690158610616982775<p>ChemoImmunoModulation: immune regulation by the antineoplastic chemotherapeutic agents</p>ShurinMRNaiditchHGutkinDWUmanskyVShurinGVCurr Med Chem2012191792180310.2174/09298671280009978522414087<p>Clinical development of immunostimulatory monoclonal antibodies and opportunities for combination</p>MeleroIGrimaldiAMPerez-GraciaJLAsciertoPAClin Cancer Res201319997100810.1158/1078-0432.CCR-12-221423460531<p>Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies</p>PeggsKSQuezadaSAChambersCAKormanAJAllisonJPJ Exp Med20092061717172510.1084/jem.20082492272217419581407<p>Immunologic and clinical effects of antibody blockade of cytotoxic T lymphocyte-associated antigen 4 in previously vaccinated cancer patients</p>HodiFSButlerMObleDASeidenMVHaluskaFGKruseAMacraeSNelsonMCanningCLowyIProc Natl Acad Sci U S A20081053005301010.1073/pnas.0712237105226857518287062<p>PD-L1 on Tumor Cells Is Induced in Ascites and Promotes Peritoneal Dissemination of Ovarian Cancer through CTL Dysfunction</p>AbikoKMandaiMHamanishiJYoshiokaYMatsumuraNBabaTYamaguchiKMurakamiRYamamotoAKharmaBClin Cancer Res2013191363137410.1158/1078-0432.CCR-12-219923340297<p>Safety, activity, and immune correlates of anti-PD-1 antibody in cancer</p>TopalianSLHodiFSBrahmerJRGettingerSNSmithDCMcDermottDFPowderlyJDCarvajalRDSosmanJAAtkinsMBLemingPDSpigelDRAntoniaSJHornLDrakeCGPardollDMChenLSharfmanWHAndersRATaubeJMMcMillerTLXuHKormanAJJure-KunkelMAgrawalSMcDonaldDKolliaGDGuptaAWiggintonJMSznolMN Engl J Med20123662624432454Epub 2012 Jun 210.1056/NEJMoa1200690354453922658127<p>Tumor-expressed B7-H1 and B7-DC in relation to PD-1+ T-cell infiltration and survival of patients with cervical carcinoma</p>KarimRJordanovaESPiersmaSJKenterGGChenLBoerJMMeliefCJvan der BurgSHClin Cancer Res2009156341634710.1158/1078-0432.CCR-09-165219825956<p>Safety and activity of anti-PD-L1 antibody in patients with advanced cancer</p>BrahmerJRTykodiSSChowLQHwuWJTopalianSLHwuPDrakeCGCamachoLHKauhJOdunsiKN Engl J Med20123662455246510.1056/NEJMoa1200694356326322658128<p>Anti-programmed death-1 and anti-programmed death-ligand 1 antibodies in cancer therapy</p>HamidOCarvajalRDExpert Opin Biol Ther2013136847861Epub 2013 Feb 19. PMID: 23421934 [PubMed - in process]10.1517/14712598.2013.77083623421934<p>A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma</p>BensonDMJrHofmeisterCCPadmanabhanSSuvannasankhaAJagannathSAbonourRBakanCAndrePEfeberaYTiollierJBlood20121204324433310.1182/blood-2012-06-43802823033266<p>A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission</p>VeyNBourhisJHBoisselNBordessouleDPrebetTCharbonnierAEtienneAAndrePRomagneFBensonDBlood20121204317432310.1182/blood-2012-06-43755823002117<p>Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity</p>LiuXShinNKoblishHKYangGWangQWangKLeffetLHansburyMJThomasBRuparMBlood20101153520353010.1182/blood-2009-09-24612420197554<p>Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells</p>SinhaPClementsVKFultonAMOstrand-RosenbergSCancer Res2007674507451310.1158/0008-5472.CAN-06-417417483367<p>COX-2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid-derived suppressor cells in mesothelioma, Celecoxib influences MDSC function</p>VeltmanJDLambersMEvan NimwegenMHendriksRWHoogstedenHCAertsJGHegmansJPBMC Cancer20101046410.1186/1471-2407-10-464293955220804550<p>Celecoxib for the prevention of colorectal adenomatous polyps</p>ArberNEagleCJSpicakJRaczIDitePHajerJZavoralMLechugaMJGerlettiPTangJN Engl J Med200635588589510.1056/NEJMoa06165216943401<p>Tumour macrophages as potential targets of bisphosphonates</p>RogersTLHolenIJ Transl Med2011917710.1186/1479-5876-9-177321518722005011<p>Zoledronic acid impairs myeloid differentiation to tumour-associated macrophages in mesothelioma</p>VeltmanJDLambersMEvan NimwegenMHendriksRWHoogstedenHCHegmansJPAertsJGBr J Cancer201010362964110.1038/sj.bjc.6605814293825720664588