logo logo
Anti-CCR4 monoclonal antibody enhances antitumor immunity by modulating tumor-infiltrating Tregs in an ovarian cancer xenograft humanized mouse model. Chang De-Kuan,Peterson Eric,Sun Jiusong,Goudie Calum,Drapkin Ronny I,Liu Joyce F,Matulonis Ursula,Zhu Quan,Marasco Wayne A Oncoimmunology Recent studies have demonstrated that regulatory T cells (Tregs) are recruited to tumor sites where they can suppress antitumor immunity. The chemokine receptor CCR4 is expressed at high levels on functional CD4CD25FoxP3 Tregs and production of the CCR4 ligand CCL22 by tumor cells and tumor-associated macrophages is associated with Treg recruitment to the tumor site. Here, we tested IgG1 and IgG4 isotypes of human anti-CCR4 mAb2-3 for their activity and capacity in a NSG mouse model bearing CCL22-secreting ovarian cancer (OvCA) xenograft to modulate Tregs and restore antitumor activity. Both mAb2-3 isotypes blocked chemoattraction of Tregs to CCL22-secreting OvCA cells. However, they differed in their mode of action with IgG1 causing Treg depletion and IgG4 blocking migration to the tumors. Primary T cells that were primed with OvCA-pulsed dendritic cells (DCs) demonstrated INFγ secretion that could be enhanced through Treg depletion by mAb2-3. Humanized mice reconstructed with allogeneic tumor-primed T cells (TP-T) were used to evaluate the restoration of OvCA immunity by depletion or blockade of Tregs with mAb2-3. We observed that IgG1 was more potent than IgG4 in inhibiting tumor growth. Mechanism studies demonstrated that mAb2-3 treatment lead to inhibition of IL-2 binding to its receptor. Further studies showed that mAb2-3 induced CD25 shedding (sCD25) from Tregs which lead to a decrease in IL-2-dependent survival. Together, the results demonstrate that mAb2-3 is an agonist antibody that can restore anti-OvCA immunity through modulation of Treg activity. 10.1080/2162402X.2015.1090075
Suppressive IL-17AFoxp3 and ex-Th17 IL-17AFoxp3 T cells are a source of tumour-associated T cells. Downs-Canner Stephanie,Berkey Sara,Delgoffe Greg M,Edwards Robert P,Curiel Tyler,Odunsi Kunle,Bartlett David L,Obermajer Nataša Nature communications Th17 and regulatory T (T) cells are integral in maintaining immune homeostasis and Th17-T imbalance is associated with inflammatory immunosuppression in cancer. Here we show that Th17 cells are a source of tumour-induced Foxp3 cells. In addition to natural (n)T and induced (i)T cells that develop from naive precursors, suppressive IL-17AFoxp3 and ex-Th17 Foxp3 cells are converted from IL-17AFoxp3 cells in tumour-bearing mice. Metabolic phenotyping of Foxp3-expressing IL-17A, ex-Th17 and iT cells demonstrates the dissociation between the metabolic fitness and the suppressive function of Foxp3-expressing T cell subsets. Although all Foxp3-expressing subsets are immunosuppressive, glycolysis is a prominent metabolic pathway exerted only by IL-17AFoxp3 cells. Transcriptome analysis and flow cytometry of IL-17AFoxp3 cells indicate that Folr4, GARP, Itgb8, Pglyrp1, Il1rl1, Itgae, TIGIT and ICOS are Th17-to-T cell transdifferentiation-associated markers. Tumour-associated Th17-to-T cell conversion identified here provides insights for targeting the dynamism of Th17-T cells in cancer immunotherapy. 10.1038/ncomms14649
Interleukin-2 administration alters the CD4+FOXP3+ T-cell pool and tumor trafficking in patients with ovarian carcinoma. Wei Shuang,Kryczek Ilona,Edwards Robert P,Zou Linhua,Szeliga Wojciech,Banerjee Mousumi,Cost Marilyn,Cheng Pui,Chang Alfred,Redman Bruce,Herberman Ronald B,Zou Weiping Cancer research Interleukin (IL)-2 is used in the immunotherapy of patients with certain cancer and HIV infection. IL-2 treatment reliably results in 16% to 20% objective clinical response rate in cancer patients, with significant durability of responses in selected patients. However, the mechanisms of therapeutic activity in responding versus nonresponding patients remain poorly understood. CD4(+)CD25(+)FOXP3(+) regulatory T (Treg) cells contribute to immunosuppressive networks in human tumors. We treated 31 ovarian cancer patients with IL-2. We show that administration of IL-2 induces the proliferation of existent Treg cells in patients with ovarian cancer. The potency of Treg cell proliferation is negatively determined by the initial prevalence of Treg cells, suggesting that Treg cells are a factor for self-controlling Treg cell proliferation. After IL-2 cessation, the number of Treg cells more efficiently dropped in clinical responders than nonresponders. Furthermore, IL-2 treatment stimulates chemokine receptor CXCR4 expression on Treg cells, enables Treg cell migration toward chemokine CXCL12 in the tumor microenvironment, and may enforce Treg cell tumor accumulation. Our findings support the concept that administration of IL-2 numerically and functionally affects the Treg cell compartment. These data provide an important insight in evaluating the clinical benefit and therapeutic prediction of IL-2 treatment in patients with cancer. 10.1158/0008-5472.CAN-07-0565
DPP4 Inhibitor Sitagliptin Enhances Lymphocyte Recruitment and Prolongs Survival in a Syngeneic Ovarian Cancer Mouse Model. Wilson Amy L,Moffitt Laura R,Wilson Kirsty L,Bilandzic Maree,Wright Mark D,Gorrell Mark D,Oehler Martin K,Plebanski Magdalena,Stephens Andrew N Cancers Immunity plays a key role in epithelial ovarian cancer (EOC) progression with a well-documented correlation between patient survival and high intratumoral CD8+ to T regulatory cell (Treg) ratios. We previously identified dysregulated DPP4 activity in EOCs as a potentially immune-disruptive influence contributing to a reduction in CXCR3-mediated T-cell infiltration in solid tumours. We therefore hypothesized that inhibition of DPP4 activity by sitagliptin, an FDA-approved inhibitor, would improve T-cell infiltration and function in a syngeneic ID8 mouse model of EOC. Daily oral sitagliptin at 50 mg/kg was provided to mice with established primary EOCs. Sitagliptin treatment decreased metastatic tumour burden and significantly increased overall survival and was associated with significant changes to the immune landscape. Sitagliptin increased overall CXCR3-mediated CD8+ T-cell trafficking to the tumour and enhanced the activation and proliferation of CD8+ T-cells in tumour tissue and the peritoneal cavity. Substantial reductions in suppressive cytokines, including CCL2, CCL17, CCL22 and IL-10, were also noted and were associated with reduced CD4+ CD25+ Foxp3+ Treg recruitment in the tumour. Combination therapy with paclitaxel, however, typical of standard-of-care for patients in palliative care, abolished CXCR3-specific T-cell recruitment stimulated by sitagliptin. Our data suggest that sitagliptin may be suitable as an adjunct therapy for patients between chemotherapy cycles as a novel approach to enhance immunity, optimise T-cell-mediated function and improve overall survival. 10.3390/cancers13030487
The expression of the regulatory T cell-specific forkhead box transcription factor FoxP3 is associated with poor prognosis in ovarian cancer. Wolf Dominik,Wolf Anna M,Rumpold Holger,Fiegl Heidi,Zeimet Alain G,Muller-Holzner Elisabeth,Deibl Martina,Gastl Guenther,Gunsilius Eberhard,Marth Christian Clinical cancer research : an official journal of the American Association for Cancer Research PURPOSE:The forkhead box transcription factor FoxP3 is specifically expressed in T cells with regulatory properties (Treg). Recently, high numbers of Treg were described to be associated with poor survival in different malignancies. The aim of the presented study was determine the prognostic effect of FoxP3 mRNA expression (reflecting the tissue content of Treg) in ovarian carcinoma and its relation with cytokines, such as IFN-gamma. EXPERIMENTAL DESIGN:Total RNA was isolated from 99 ovarian carcinoma and from 14 healthy ovarian biopsies. Real-time PCR for FoxP3 was done and correlated with IFN-gamma-, CD3-, IRF-1-, SOCS-1-, HER-2-, and iNOS expression as well as patients' outcome. The mRNA data was corroborated by FoxP3 immunohistochemistry. RESULTS:Quantitation of FoxP3 expression identified a patient subgroup (>81th percentile), which is characterized by a significantly worse prognosis in terms of overall survival (27.8 versus 77.3 months, P = 0.0034) and progression-free survival (18 versus 57.5 months; P = 0.0041). FoxP3 expression correlated with IFN-gamma, IRF-1, and CD3 expression. High FoxP3 expression represents an independent prognostic factor for overall survival (P = 0.004) and progression-free survival (P = 0.004). CONCLUSIONS:High expression levels of FoxP3 might represent a surrogate marker for an immunosuppressive milieu contributing to tumor immune escape. Strategies selectively depleting Treg might improve the antitumor activity of endogenously arising tumor-reactive T cells and immunotherapies using vaccines or antibodies. 10.1158/1078-0432.CCR-05-1244
CXCR3+ T regulatory cells selectively accumulate in human ovarian carcinomas to limit type I immunity. Redjimi Nassima,Raffin Caroline,Raimbaud Isabelle,Pignon Pascale,Matsuzaki Junko,Odunsi Kunle,Valmori Danila,Ayyoub Maha Cancer research Antitumor type I T-cell responses involving IFN-γ production are critical to control cancer, but the efficacy of this response is limited by a variety of immunosuppressive mechanisms that promote tumoral immune escape. One critical mechanism involves the accumulation of FOXP3(+) T regulatory cells (Treg), a class of suppressive T cells that prevent excessive tissue destruction caused by unchecked immune responses. Recent studies have revealed that FOXP3(+) Treg include distinct subsets specifically controlling over the corresponding effector subset. In particular, CXCR3(+) Treg have been described as a subset specialized in the control of type I T-cell responses in vivo. Here, we show that CXCR3(+) Treg are highly enriched in human ovarian carcinomas, particularly in solid tumor masses, where they represent the majority of Treg. Tumor-associated CXCR3(+.) Treg coexpress T-bet but do not secrete IFN-γ ex vivo and suppress proliferation and IFN-γ secretion of T effectors. In addition, they coexpress Helios, suggesting that they originate from natural Treg. Finally, we show that the proportion of CXCR3(+) Treg at tumor sites is directly correlated with that of CXCR3(+) T effectors, consistent with expression of CXCR3 ligands. Together, our findings support the concept that natural CXCR3(+) T-bet(+) Treg selectively accumulate in ovarian tumors to control type I T-cell responses, resulting in the collateral limitation of efficient antitumor immunity. 10.1158/0008-5472.CAN-12-0579
FoxP3+ regulatory T cells in peripheral blood of patients with epithelial ovarian cancer. Erfani Nasrollah,Hamedi-Shahraki Mahboobeh,Rezaeifard Somayeh,Haghshenas Mohammadreza,Rasouli Manoochehr,Samsami Dehaghani Alamtaj Iranian journal of immunology : IJI BACKGROUND:Ovarian cancer is the fifth leading cause of death from malignancy in women. CD4+CD25+FoxP3+ regulatory T (Treg) cells are a subset of T lymphocytes with great inhibitory impact on immune response. OBJECTIVES:To investigate the percentage of CD4+CD25+FoxP3+ regulatory T cells in the peripheral blood of the Iranian patients with epithelial ovarian cancer compared to healthy women and to evaluate the correlation of the Treg cell percentage with clinicopathological characteristics including cancer stage and CA-125 serum level. METHODS:Seventeen women with epithelial ovarian cancer and 20 healthy subjects were enrolled in the study. Peripheral blood mononuclear cells were stained at the surface, for CD4 and CD25 molecules, followed by fixation, permeabilization and intracellular staining for FoxP3 molecule. After processing and flowcytometry analysis, prevalence of Treg cells was determined as the percentages of CD25+FoxP3+ cells among CD4+ lymphocytes. RESULTS:Despite no difference in the percentage of total CD4+ lymphocytes, analysis indicated that Treg cell percentage was significantly higher in ovarian cancer patients than controls (5.7 ± 3.1% versus 2.8 ± 1.4%, p=0.002). A trend toward higher Treg cells was observed in higher stages of ovarian cancer (III+IV) in comparison to lower stages (I+II) (6.5 ± 3.2% vs. 4.44 ± 2.7%, p=0.2). Higher percentage of Treg cells was also observed in the patients with high CA125 (CA-125 >100 U/mL) in comparison to those with low CA-125 serum level (CA-125 ≤ 100 U/mL) although the difference was not significant (6.44 versus 4.18%, p=0.19). CONCLUSION:Increased frequency of Tregs in ovarian cancer might participate in immune suppression in these patients. The findings collectively suggest the likely impact of Treg cell-targeted immunotherapy in ovarian cancer. IJIv11i2A5
Analysis of CD8+ Treg cells in patients with ovarian cancer: a possible mechanism for immune impairment. Zhang Shuping,Ke Xing,Zeng Suyun,Wu Meng,Lou Jianfang,Wu Lei,Huang Peijun,Huang Lei,Wang Fang,Pan Shiyang Cellular & molecular immunology Regulatory T (Treg) cells may participate in mediating a suppressive microenvironment that blunts successful anti-tumor immunotherapy. Recent studies show that CD8(+) Treg cells might impede effective immune responses to established tumors. However, there is limited research regarding CD8(+) Treg cells in ovarian cancer (OC) patients. Here, we investigated CD8(+) Treg cells in OC patients and their in vitro induction. The immunohistochemistry of tumor-infiltrating lymphocytes revealed a significant correlation between the intratumoral CD8(+) T cells and the forkhead box p3 (Foxp3)(+) cells in the intraepithelial and stromal areas of advanced OC tissues. We examined the expression of Treg markers in CD8(+) T cells from the peripheral blood and fresh tumor tissues of OC patients using flow cytometry. Our results indicated an increase in the CD8(+) Treg cell subsets of OC patients compared with those in patients with benign ovarian tumors and healthy controls, including an increased expression of CD25, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and Foxp3 and decreased CD28 expression. To demonstrate whether the tumor microenvironment could convert CD8(+) effector T cells into suppressor cells, we used an in vitro transwell culturing system. Compared with the CD8(+) T cells cultured alone, the CD8(+) Treg cells induced in vitro by coculture with SK-OV-3/A2780 showed increased CTLA-4 and Foxp3 expression and decreased CD28 expression. In addition, the in vitro-induced CD8(+) Treg cells inhibited naı¨ve CD4(+) T-cell proliferation, which was partially mediated through TGF-β1 and IFN-γ. Our study suggests that CD8(+) Treg cells were increased in OC patients and could be induced in vitro, which may be the way that tumors limit antitumor immunity and evade immune surveillance. 10.1038/cmi.2015.57
Regulatory T cells, inherited variation, and clinical outcome in epithelial ovarian cancer. Knutson Keith L,Maurer Matthew J,Preston Claudia C,Moysich Kirsten B,Goergen Krista,Hawthorne Kieran M,Cunningham Julie M,Odunsi Kunle,Hartmann Lynn C,Kalli Kimberly R,Oberg Ann L,Goode Ellen L Cancer immunology, immunotherapy : CII The immune system constitutes one of the host factors modifying outcomes in ovarian cancer. Regulatory T cells (Tregs) are believed to be a major factor in preventing the immune response from destroying ovarian cancers. Understanding mechanisms that regulate Tregs in the tumor microenvironment could lead to the identification of novel targets aimed at reducing their influence. In this study, we used immunofluorescence-based microscopy to enumerate Tregs, total CD4 T cells, and CD8(+) cytotoxic T cells in fresh frozen tumors from over 400 patients with ovarian cancer (>80 % high-grade serous). We sought to determine whether Tregs were associated with survival and genetic variation in 79 genes known to influence Treg induction, trafficking, or function. We used Cox regression, accounting for known prognostic factors, to estimate hazard ratios (HRs) associated with T cell counts and ratios. We found that the ratios of CD8 T cells and total CD4 T cells to Tregs were associated with improved overall survival (CD8/Treg HR 0.84, p = 0.0089; CD4/Treg HR 0.88, p = 0.046) and with genetic variation in IL-10 (p = 0.0073 and 0.01, respectively). In multivariate analyses, the associations between the ratios and overall survival remained similar (IL-10 and clinical covariate-adjusted CD8/Treg HR 0.85, p = 0.031; CD4/Treg HR 0.87, p = 0.093), suggesting that this association was not driven by variation in IL-10. Thus, integration of novel tumor phenotyping measures with extensive clinical and genetic information suggests that the ratio of T cells to Tregs may be prognostic of outcome in ovarian cancer, regardless of inherited genotype in genes related to Tregs. 10.1007/s00262-015-1753-x
Changes in regulatory T cells in patients with ovarian cancer undergoing surgery: Preliminary results. Wu Meng,Chen Xian,Lou Jianfang,Zhang Shuping,Zhang Xiaojie,Huang Lei,Sun Ruihong,Huang Peijun,Pan Shiyang,Wang Fang International immunopharmacology Regulatory T cells (Treg) suppress immune responses in patients with cancer. Surgery is the most effective therapeutic strategy for ovarian cancer (OC). However, the interplay between the Treg population and surgical resection remains unclear. 61 patients with OC who received no prior treatment were enrolled in the study. Treg percentages were characterized from peripheral blood mononuclear cells. We investigated CD4CD25, CD4CD25Foxp3, CD8CD28, and CD8Foxp3 Tregs in OC patients and their postoperative changes using flow cytometry. Treg percentages were significantly higher in OC patients than those in benign ovarian tumors (BOT) and healthy controls. Higher percentages of Tregs were found in patients with stage III/IV than stage I/II OC. Treg percentages were significantly decreased postoperatively. The postoperative Treg percentages in patients with stage I/II OC were similar to those in BOT patients, while postoperative Treg percentages in patients with stage III/IV OC remained higher. Tregs were markedly lower on postoperative day (POD) 3 than preoperatively. They increased slightly after 7days, but remained lower than preoperative levels. These data suggested that Tregs continued to decline from POD 7 to POD 30. Treg percentages are correlated with the tumor burden and could be a key factor in monitoring the immunological status of patients with OC. 10.1016/j.intimp.2017.04.004
Regulatory T Cells in Ovarian Cancer Are Characterized by a Highly Activated Phenotype Distinct from that in Melanoma. Toker Aras,Nguyen Linh T,Stone Simone C,Yang S Y Cindy,Katz Sarah Rachel,Shaw Patricia A,Clarke Blaise A,Ghazarian Danny,Al-Habeeb Ayman,Easson Alexandra,Leong Wey L,McCready David R,Reedijk Michael,Guidos Cynthia J,Pugh Trevor J,Bernardini Marcus Q,Ohashi Pamela S Clinical cancer research : an official journal of the American Association for Cancer Research Regulatory T (Treg) cells expressing the transcription factor FOXP3 are essential for the maintenance of immunologic self-tolerance but play a detrimental role in most cancers due to their ability to suppress antitumor immunity. The phenotype of human circulating Treg cells has been extensively studied, but less is known about tumor-infiltrating Treg cells. We studied the phenotype and function of tumor-infiltrating Treg cells in ovarian cancer and melanoma to identify potential Treg cell-associated molecules that can be targeted by tumor immunotherapies. The phenotype of intratumoral and circulating Treg cells was analyzed by multicolor flow cytometry, mass cytometry, RNA-seq, and functional assays. Treg cells isolated from ovarian tumors displayed a distinct cell surface phenotype with increased expression of a number of receptors associated with TCR engagement, including PD-1, 4-1BB, and ICOS. Higher PD-1 and 4-1BB expression was associated with increased responsiveness to further TCR stimulation and increased suppressive capacity, respectively. Transcriptomic and mass cytometry analyses revealed the presence of Treg cell subpopulations and further supported a highly activated state specifically in ovarian tumors. In comparison, Treg cells infiltrating melanomas displayed lower FOXP3, PD-1, 4-1BB, and ICOS expression and were less potent suppressors of CD8 T-cell proliferation. The highly activated phenotype of ovarian tumor-infiltrating Treg cells may be a key component of an immunosuppressive tumor microenvironment. Receptors that are expressed by tumor-infiltrating Treg cells could be exploited for the design of novel combination tumor immunotherapies. . 10.1158/1078-0432.CCR-18-0554
TGF-β1 contributes to CD8+ Treg induction through p38 MAPK signaling in ovarian cancer microenvironment. Wu Meng,Chen Xian,Lou Jianfang,Zhang Shuping,Zhang Xiaojie,Huang Lei,Sun Ruihong,Huang Peijun,Wang Fang,Pan Shiyang Oncotarget CD8+ regulatory T cells (Tregs) contribute to cancer progression and immune evasion. We previously reported that CD8+ Tregs could be induced in vitro by co-culture of CD8+ T cells with the OC cell lines SKOV3/A2780. Here, we described the role of TGF-β1 in CD8+ Treg induction by the OC microenvironment. OC patients expressed high levels of TGF-β1, as did the co-culture supernatant from CD8+ T cells and SKOV3. Additionally, TGF-β1 levels were positively correlated with CD8+ Treg percentages in OC. Neutralization experiments, cytokine studies and proliferation assays revealed that the in vitro-induced CD8+Tregs depended at least partially on up-regulated expression of TGF-β1 to exert their suppressive function. CD8+ T cells cultured with SKOV3 exhibited marked activation of p38 MAPK than CD8+ T cells cultured alone, which could be inhibited by TGF-β1-neutralizing antibody. Moreover, the p38 specific inhibitor SB203580 dose-dependently blocked the TGF-β1 activated conversion of CD8+ T cells into CD8+ Tregs. These data suggested that in vitro-induction of CD8+ Tregs depended in part on TGF-β1 activation of p38 MAPK signaling. Therefore, p38 MAPK could be a therapeutic target in OC anti-tumor immunotherapy. 10.18632/oncotarget.10003
The possible regulatory effect of the PD-1/PD-L1 signaling pathway on Tregs in ovarian cancer. Chen Jian-Xia,Yi Xi-Juan,Gao Shan-Xia,Sun Jin-Xia General physiology and biophysics Aim of this study was to investigate the possible regulatory effect of the programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) signaling pathway on Tregs in ovarian cancer. Immunohistochemistry was used to detect the expression of PD-L1 and PD-1 and the presence of FOXP3+ Tregs in ovarian cancer. Then, ovarian cancer HO8910 cells were subjected to transfection with PD-L1 siRNA in vitro. CCK-8, Transwell and wound healing assays were performed to detect the biological behaviors of ovarian cancer cells. Human T-cells isolated from human peripheral blood were cocultured with HO8910 cells, which were divided into the Control, TGF-β, and TGF-β+ anti-PD-L1 groups. The proportion of differentiated Tregs was detected by flow cytometry. Mouse models of ovarian cancer were established, and PD-L1 antibody therapy was administered. Tumor growth and Treg recruitment were observed. PD-L1, PD-1 and FOXP3+ Tregs were found in ovarian cancer tissue. Patients with tumors with an advanced stage and low differentiation and lymph node metastasis had significantly higher levels of PD-1, PD-L1 and FOXP3+ Tregs. After transfection with PD-L1 siRNA, HO8910 cells showed a significant reduction in PD-L1 expression, proliferation, migration and invasion. After T-cells were cocultured with ovarian cancer cells, the TGF-β+ anti-PD-L1 group showed a substantial decline in the differentiation of T-cells into Tregs compared with the TGF-β group. Moreover, mice in the anti-PD-L1 group had significantly reduced tumor growth rates, Treg proportions in the tumor microenvironment, and FOXP3 expression. 10.4149/gpb_2020011
Interaction between Treg cells and tumor-associated macrophages in the tumor microenvironment of epithelial ovarian cancer. Zhu Qinyi,Wu Xiaoli,Wu Yueqian,Wang Xipeng Oncology reports Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. Inflammatory cells in the EOC microenvironment play a key role in tumor progression. In the present study, we investigated the mechanism of the accumulation of regulatory T cells (Tregs) induced by interleukin-10 (IL-10) derived from tumor-associated macrophages (TAMs) in the EOC microenvironment. The frequency of Tregs and TAMs was detected by immunofluorescence in 40 EOC tissues and 20 benign ovarian tumors, as well as the expression of IL-10 which was assessed by immunohistochemistry. It was found that the frequency of Treg cells and TAMs was significantly higher in the EOC than those in the benign ovarian tumors. The expression of IL-10 was also found to be higher in the EOC than that in the benign tumors. EOC patients with a high frequency of Tregs exhibited a significantly shorter overall survival time compared to those with a low frequency of Tregs. In addition, the expression of IL-10 in ascites and blood serum and the IL-10 released in the co-cultured system supernatants were detected by ELISA. Following CD4+ T-cell co-culturing with macrophages and IL-10, it was observed by flow cytometric analysis that the frequency of Treg cells was increased in the presence of IL-10. It was also established that IL-10 released in the co-cultured supernatants was increased. We also detected the mechanism of Treg cells induced by IL-10 in vivo. The SKOV3 cell tumor volume and weight were much higher in the presence of IL-10 in a mouse subcutaneous model. These data suggest that IL-10 secreted by TAMs increase the frequency of Treg cells through the activation of Foxp3 during T-cell differentiation and promotes tumor progression. 10.3892/or.2016.5136
Effects of Treg cells and IDO on human epithelial ovarian cancer cells under hypoxic conditions. Liu Jun,Zhang Haiyan,Jia Luoqi,Sun Hong Molecular medicine reports The aim of the present study was to explore the effect of hypoxia on ovarian cancer. A total of 6 samples were analyzed: SKOV3‑IP cells (ovarian cancer cell line); SKOV3‑IP and regulatory T (Treg) cells; SKOV3‑IP and cytotoxic T lymphocytes (CTLs); SKOV3‑IP and natural killer (NK) cells; SKOV3‑IP co-cultured with CTLs and Treg cells; and SKOV3‑IP co-cultured with Treg cells and NK cells. The expression of indoleamine 2,3‑dioxygenase (IDO) was detected by reverse transcription-polymerase chain reaction (RT‑PCR) and western blot analysis. An enzyme‑linked immunosorbent assay (ELISA) was used to detect the concentration of transforming growth factor‑β (TGF‑β), interferon‑γ (IFN‑γ), interleukin‑2 (IL‑2), interleukin‑10 (IL‑10), and perforin. Moreover, ovarian cancer cell apoptosis and invasive ability were examined using flow cytometry and a Transwell chamber assay. IDO expression was significantly reduced in hypoxia and enhanced by Treg cells. Treg cells inhibited the IL‑2, IFN‑γ and perforin secretion, and significantly (P<0.05) increased the IL‑10 and TGF‑β levels. The effects of Treg cells were enhanced with prolongation of the cell exposure to hypoxic conditions. In addition, Treg cells attenuated the promotive effect of CTLs and NK cells on cancer cell apoptosis. In addition, Treg cells significantly increased the SKOV3‑IP invasive ability (P=0.00109) under hypoxic conditions. Our results suggest that IDO and Treg cells may serve as important therapeutic targets for patients with ovarian cancer. 10.3892/mmr.2014.2893
Exosomes Released from Tumor-Associated Macrophages Transfer miRNAs That Induce a Treg/Th17 Cell Imbalance in Epithelial Ovarian Cancer. Zhou Jieru,Li Xiaoduan,Wu Xiaoli,Zhang Ting,Zhu Qinyi,Wang Xinjing,Wang Husheng,Wang Kai,Lin Yingying,Wang Xipeng Cancer immunology research The immune microenvironment is crucial for epithelial ovarian cancer (EOC) progression and consists of tumor-associated macrophages (TAM) and T lymphocytes, such as regulatory T cells (Treg) and T helper 17 (Th17) cells. In this study, the Treg/Th17 ratio was significantly higher in EOC and in metastatic peritoneal tissues than in benign ovarian tumors and benign peritoneum. The Treg/Th17 ratio was associated with histologic grade and was an independent prognostic factor for overall survival of EOC patients. On the basis of microarray analysis of exosomes derived from TAMs, we identified miRNAs enriched in the exosomes, including miR-29a-3p and miR-21-5p. When the two miRNA mimics were transfected into CD4 T cells, they directly suppressed STAT3 and regulated Treg/Th17 cells, inducing an imbalance, and they had a synergistic effect on STAT3 inhibition. Taken together, these results indicate that exosomes mediate the interaction between TAMs and T cells, generating an immune-suppressive microenvironment that facilitates EOC progression and metastasis. These findings suggest that targeting these exosomes or their associated miRNAs might pave the way for the development of novel treatments for EOC. 10.1158/2326-6066.CIR-17-0479
Analysis of the treg cell population in the peripheral blood of ovarian cancer patients in relation to the long-term outcomes. Dutsch-Wicherek Magdalena Maria,Szubert Sebastian,Dziobek Konrad,Wisniewski Michal,Lukaszewska Ewelina,Wicherek Lukasz,Jozwicki Wojciech,Rokita Wojciech,Koper Krzysztof Ginekologia polska OBJECTIVES:There is growing evidence that Treg cell infiltration into the cancer nest is associated with poor prognosis. How- ever, the Treg cell population in the peripheral blood may change when a different type of anticancer therapy is applied. Since Treg cells may support tumor growth by enhancing the suppressive profile of the cancer microenvironment, the assessment of Treg cells can bring to light important information regarding prognosis. Thus we decided to analyze the Treg cell population in the peripheral blood in relation to long-term outcomes in the group of patients with ovarian cancer. MATERIAL AND METHODS:The 80 patients included in the study were treated surgically followed by chemiotherapy for ovar- ian cancer between October 2010 through May 2011.The peripheral blood samples from the patients were collected directly prior to chemotherapy. Information on any patients who died was retrieved from the database of the Cuiavia-Pomerania Regional Office of the National Health System of Poland. CD4+CD25+FOXP3+ lymphocytes T were assed by flow cytometry. We have analyzed the long term outcomes of treatment regarding to the level of Treg cells in peripheral blood. RESULTS:We found that patients with serous adenocarcinomas had significantly higher Treg levels compared to those patients with non-serous types. Patients who had a higher percentage of Treg cells within the CD4+ cell population prior to the beginning of the treatment had worse long-term outcomes from the applied therapy. CONCLUSIONS:The assessment of Treg levels prior to the start of chemotherapy is clinically useful and may predict overall survival in ovarian cancer patients. 10.5603/GP.2019.0032
The Ovarian Cancer Chemokine Landscape Is Conducive to Homing of Vaccine-Primed and CD3/CD28-Costimulated T Cells Prepared for Adoptive Therapy. Zsiros Emese,Duttagupta Priyanka,Dangaj Denarda,Li Hongzhe,Frank Renee,Garrabrant Thomas,Hagemann Ian S,Levine Bruce L,June Carl H,Zhang Lin,Wang Ena,Marincola Francesco M,Bedognetti Davide,Powell Daniel J,Tanyi Janos,Feldman Michael D,Kandalaft Lana E,Coukos George Clinical cancer research : an official journal of the American Association for Cancer Research PURPOSE:Chemokines are implicated in T-cell trafficking. We mapped the chemokine landscape in advanced stage ovarian cancer and characterized the expression of cognate receptors in autologous dendritic cell (DC)-vaccine primed T cells in the context of cell-based immunotherapy. EXPERIMENTAL DESIGN:The expression of all known human chemokines in patients with primary ovarian cancer was analyzed on two independent microarray datasets and validated on tissue microarray. Peripheral blood T cells from five HLA-A2 patients with recurrent ovarian cancer, who previously received autologous tumor DC vaccine, underwent CD3/CD28 costimulation and expansion ex vivo. Tumor-specific T cells were identified by HER2/neu pentamer staining and were evaluated for the expression and functionality of chemokine receptors important for homing to ovarian cancer. RESULTS:The chemokine landscape of ovarian cancer is heterogeneous with high expression of known lymphocyte-recruiting chemokines (CCL2, CCL4, and CCL5) in tumors with intraepithelial T cells, whereas CXCL10, CXCL12, and CXCL16 are expressed quasi-universally, including in tumors lacking tumor-infiltrating T cells. DC-vaccine primed T cells were found to express the cognate receptors for the above chemokines. Ex vivo CD3/CD28 costimulation and expansion of vaccine-primed Tcells upregulated CXCR3 and CXCR4, and enhanced their migration toward universally expressed chemokines in ovarian cancer. CONCLUSIONS:DC-primed tumor-specific T cells are armed with the appropriate receptors to migrate toward universal ovarian cancer chemokines, and these receptors are further upregulated by ex vivo CD3/CD28 costimulation, which render T cells more fit for migrating toward these chemokines. Clin Cancer Res; 21(12); 2840-50. ©2015 AACR. 10.1158/1078-0432.CCR-14-2777
Ovarian cancer stem cells promote tumour immune privilege and invasion via CCL5 and regulatory T cells. You Y,Li Y,Li M,Lei M,Wu M,Qu Y,Yuan Y,Chen T,Jiang H Clinical and experimental immunology Emerging evidence indicates a link between the increased proportion of regulatory T cells (T ) and reduced survival in patients who have been diagnosed with cancer. Cancer stem cells (CSCs) have been indicated to play a vital role in tumour initiation, drug resistance and recurrence. However, the relationship between T and CSCs remains largely unknown. Here, we sorted out ovarian cancer stem-like side population (SP) cells and CD133 cells to investigate the influence of ovarian CSCs on T . Among the various immune-related molecules that we assessed, C-C motif chemokine ligand 5 (CCL5) was the most elevated in ovarian CSCs relative to that in the non-CSCs. The expression of its receptor, C-C motif chemokine receptor 5 (CCR5), was also increased on the surface of T in ovarian cancer patients. This receptor-ligand expression profile indicated that ovarian CSCs recruit T via CCL5-CCR5 interactions. We further assessed the expression of interleukin (IL)-10 in T cultured with different cancer cells. T cultured in conditioned medium (CM) from ovarian CD133 cells expressed a higher level of IL-10 than T cultured in CM from CD133 cells, indicating that T exert pronounced immune-inhibitory functions in CSC-rich environments. Furthermore, co-culture with ovarian cancer cell lines induced the expression of matrix metalloproteinase-9 (MMP9) in T which, in turn, enhanced the degradation of the extracellular matrix and enabled the invasion of tumour cells, thereby facilitating tumour metastasis. For the first time, to our knowledge, our findings describe the relationship between ovarian CSCs and T , and demonstrated that these two cell populations co-operate to promote tumour immune tolerance and enhance tumour progression. 10.1111/cei.13044
Integrated digital pathology and transcriptome analysis identifies molecular mediators of T-cell exclusion in ovarian cancer. Desbois Mélanie,Udyavar Akshata R,Ryner Lisa,Kozlowski Cleopatra,Guan Yinghui,Dürrbaum Milena,Lu Shan,Fortin Jean-Philippe,Koeppen Hartmut,Ziai James,Chang Ching-Wei,Keerthivasan Shilpa,Plante Marie,Bourgon Richard,Bais Carlos,Hegde Priti,Daemen Anneleen,Turley Shannon,Wang Yulei Nature communications Close proximity between cytotoxic T lymphocytes and tumour cells is required for effective immunotherapy. However, what controls the spatial distribution of T cells in the tumour microenvironment is not well understood. Here we couple digital pathology and transcriptome analysis on a large ovarian tumour cohort and develop a machine learning approach to molecularly classify and characterize tumour-immune phenotypes. Our study identifies two important hallmarks characterizing T cell excluded tumours: 1) loss of antigen presentation on tumour cells and 2) upregulation of TGFβ and activated stroma. Furthermore, we identify TGFβ as an important mediator of T cell exclusion. TGFβ reduces MHC-I expression in ovarian cancer cells in vitro. TGFβ also activates fibroblasts and induces extracellular matrix production as a potential physical barrier to hinder T cell infiltration. Our findings indicate that targeting TGFβ might be a promising strategy to overcome T cell exclusion and improve clinical benefits of cancer immunotherapy. 10.1038/s41467-020-19408-2
Epigenetic Attire in Ovarian Cancer: The Emperor's New Clothes. Cancer research Ovarian cancer is an aggressive epithelial tumor that remains a major cause of cancer morbidity and mortality in women. Epigenetic alterations including DNA methylation and histone modifications are being characterized in ovarian cancer and have been functionally linked to processes involved in tumor initiation, chemotherapy resistance, cancer stem cell survival, and tumor metastasis. The epigenetic traits of cancer cells and of associated tumor microenvironment components have been shown to promote an immunosuppressive tumor milieu. However, DNA methylation and histone modifications are reversible, and therapies targeting the epigenome have been implicated in potential reinvigoration of the antitumor immunity. In this review, we provide an overview specifically of DNA methylation and histone modifications as "clothes of the ovarian cancer genome" in relationship to their functional effects and highlight recent developments in the field. We also address the clinical implications of therapeutic strategies to remove or alter specific articles of genomic "clothing" and restore normal cellular function. As the clothes of the genome continue to be deciphered, we envision that the epigenome will become an important therapeutic target for cancer. 10.1158/0008-5472.CAN-19-3837
MiR-217 Inhibits M2-Like Macrophage Polarization by Suppressing Secretion of Interleukin-6 in Ovarian Cancer. Jiang Bin,Zhu Shu-Juan,Xiao Song-Shu,Xue Min Inflammation Ovarian cancer is one of the most deadly cancers with rapid proliferation and poor prognosis among patients. Therapies focusing on regulation of tumor immunity and microenvironments are developing. MiR-217 was dysregulated in cancer progress and plays important roles in tumorigenesis and metastasis. However, the role of miR-217 in regulation of macrophage polarization and its underlying molecular mechanism remain unclear. The expression of miR-217 in ovarian cancerous tissues and cell lines were assessed by qRT-PCR. And we detected the staining of CD86 and CD206 via flow-cytometry and the levels of Arg-1 and CCR2 by western-blot in order to evaluate M2 macrophage polarization. The targeting regulation of miR-217 on pro-inflammatory factor IL-6 was assessed by dual-luciferase reporter assay and western-blot. ELISA assay was used to evaluate the secretion of IL-6 and IL-10 of cells. MiR-217 was found to be downregulated in ovarian cancerous tissues and cell lines. This downregulation correlated with an increased expression of the IL-6, Arg-1, CCR2, and CD206 gene. The overexpression of miR-217 in SKOV3 cells can inhibit the polarization of macrophages towards an M2-like phenotype. We also found that IL-6 was validated to induce M2 macrophage polarization and its secretion in SKOV-3 cells was inhibited by miR-217 directly. Moreover, we revealed that miR-217 suppressed M2 macrophage polarization partly thought JAK/STAT3 signal pathway. Taken together, these findings indicate that miR-217 inhibits tumor-induced M2 macrophage polarization through targeting of IL-6 and regulation JAK3/STAT3 signaling pathway, which may provide a potential therapeutic target for treating ovarian cancer. 10.1007/s10753-019-01004-2
DFMO and 5-Azacytidine Increase M1 Macrophages in the Tumor Microenvironment of Murine Ovarian Cancer. Travers Meghan,Brown Stephen M,Dunworth Matthew,Holbert Cassandra E,Wiehagen Karla R,Bachman Kurtis E,Foley Jackson R,Stone Meredith L,Baylin Stephen B,Casero Robert A,Zahnow Cynthia A Cancer research Although ovarian cancer has a low incidence rate, it remains the most deadly gynecologic malignancy. Previous work has demonstrated that the DNMTi 5-Azacytidine (5AZA-C) activates type I interferon signaling to increase IFNγ T cells and natural killer (NK) cells and reduce the percentage of macrophages in the tumor microenvironment. To improve the efficacy of epigenetic therapy, we hypothesized that the addition of α-difluoromethylornithine (DFMO), an ornithine decarboxylase inhibitor, may further decrease immunosuppressive cell populations improving outcome. We tested this hypothesis in an immunocompetent mouse model for ovarian cancer and found that , 5AZA-C and DFMO, either alone or in combination, significantly increased survival, decreased tumor burden, and caused recruitment of activated (IFNγ) CD4 T cells, CD8 T cells, and NK cells. The combination therapy had a striking increase in survival when compared with single-agent treatment, despite a smaller difference in recruited lymphocytes. Instead, combination therapy led to a significant decrease in immunosuppressive cells such as M2 polarized macrophages and an increase in tumor-killing M1 macrophages. In this model, depletion of macrophages with a CSF1R-blocking antibody reduced the efficacy of 5AZA-C + DFMO treatment and resulted in fewer M1 macrophages in the tumor microenvironment. These observations suggest our novel combination therapy modifies macrophage polarization in the tumor microenvironment, recruiting M1 macrophages and prolonging survival. SIGNIFICANCE: Combined epigenetic and polyamine-reducing therapy stimulates M1 macrophage polarization in the tumor microenvironment of an ovarian cancer mouse model, resulting in decreased tumor burden and prolonged survival. 10.1158/0008-5472.CAN-18-4018
Soluble forms of immune checkpoints in ovarian cancer. Kovaleva O V,Belova T P,Kushlinsky D N,Korotkova E A,Podlesnaya P A,Gratchev A N,Zinoviev S V,Tereshkina I V,Sokolov N Yu,Kudlay D A,Kushlinskii N E Klinicheskaia laboratornaia diagnostika The data of a complex immunoassay comparative study of the content of soluble forms of sPD-1, sPD-L1, sNKG2D, sNKG2DL1, sB7-H3 and sHLA-G in the blood plasma of 75 patients with epithelial ovarian cancer and 20 healthy donors of the control group are presented. The diagnostic significance of the studied proteins was determined. The study showed that the profile of soluble immunity checkpoints differs when malignant ovarian pathology occurs. There was a statistically significant decrease in the content of sPD-L1, sNKG2DL1, sB7-H3, and sHLA-G in the blood plasma of patients compared with the control group. Differences were found in the content of the studied markers depending on the histological type of tumors. Correlations between the soluble forms of some of the studied proteins are shown, indicating the presence of independent mechanisms of immune regulation in ovarian cancer, which may explain the insufficient effectiveness of the existing immunotherapy for this type of tumor. The results obtained will undoubtedly facilitate the development of new effective methods for the diagnostics and therapy of ovarian cancer. 10.51620/0869-2084-2021-66-2-80-86
Metabolic factors contribute to T-cell inhibition in the ovarian cancer ascites. Gong Yueqing,Yang Jianling,Wang Yan,Xue Lixiang,Wang Junjie International journal of cancer Malignant ascites is one of the major clinical features of ovarian cancer, which serves as a carrier for the peritoneal dissemination of tumor cells and predicts a poor prognosis in patients. In the microenvironment of ovarian cancer ascites, antitumor immunity is suppressed, which enables the tumor cells to escape from immune surveillance. The metabolic factors, including hypoxia, nutrient deprivation and accumulation of metabolic products, contribute to the immunosuppressive status of malignant ascites. The malignant ascites and ovarian solid tumors exhibit differential metabolic profiles. In this review, we have summarized the most recent findings on the interaction between immune cells and metabolic factors in the ovarian cancer ascites. The effects of metabolic factors on the antitumor functions of T-cells in the malignant ascites were analyzed. Finally, we have discussed the potential directions for future research in this field. 10.1002/ijc.32990
Immunogenomic Analyses of Advanced Serous Ovarian Cancer Reveal Immune Score is a Strong Prognostic Factor and an Indicator of Chemosensitivity. Hao Dapeng,Liu Jie,Chen Meng,Li JingJing,Wang Li,Li Xiaobo,Zhao Qi,Di Li-Jun Clinical cancer research : an official journal of the American Association for Cancer Research Ovarian cancer is one of the first human cancers for which immune response was reported to be important for the clinical outcome. To elucidate the mechanistic relationship between immune repertoire and cancer genotype in ovarian cancer, the development of a well-defined immune score for ovarian cancer is required. From a collection of 2,203 patient samples of advanced ovarian cancer from public available resources, we evaluated the prognostic values for a compendium of immune marker genes and proposed an immune score. The relationships between immune score, tumor-infiltrating immune cells, cancer genotypes, and their impact on patient outcome were characterized. Loss of chemokine and IFNγ pathway genes is frequent in ovarian cancer and is significantly associated with low immune score and poor outcome. Chemotherapy can increase the immune score of tumors by inducing the expression of IFNγ inducible chemokines. High immune score is significantly associated with BRCA1/2 mutation status and the response to chemotherapy. Multivariate analysis revealed that immune score is a strong predictor of patient survival and the response to immunotherapy. Our results reveal the drivers of the immune repertoire of advanced ovarian cancer and demonstrate the importance of immune score as an independent prognostic signature and a potent indicator of intratumoral immune status. . 10.1158/1078-0432.CCR-17-3862
Effects of different degrees of depression on inflammatory response and immune function in patients with ovarian cancer. Su J P,Liu H F,Zhang H L,He Y J,Nie Y Journal of biological regulators and homeostatic agents The aim of this study was to explore the effect of depression of different degrees on inflammatory response and immune function in patients with ovarian cancer. One hundred and eight cases of ovarian cancer according to the Federation Internationale of Gynecologie and Obstetrigue (FIGO) stage II~III who visited the Gynecology Department of Affiliated HongQi Hospital of MuDanJiang Medical University between September 2015 and May 2017 were enrolled in the study. After being hospitalized, they were divided into two groups according to their Beck Depression Inventory (BDI) scores. The total score of BDI is 63, with 0~4 for the normal group (25 cases), 5~13 for the mild depression group (24 cases), 14~20 for the moderate depression group (28 cases), and 21~63 for the severe depression group (31 cases). The immune function, inflammatory reaction, tumor markers [CA125, human epididymis protein-4 (HE4), insulin-like growth factor-I (IGF-I)], platelet technology and D-dimer index were compared between the four groups. The results showed that there were different levels of depression in patients with ovarian cancer in II~III stage, and the degree of depression could stimulate the level of serum-6, and TNF -α in serum increased. The proportion of CD3+, CD4+ and NK cells in patients with severe depression decreased, and their immunity also decreased. Depression increased the levels of CA125, HE4 and IGF-I in serum and ascites of ovarian cancer patients, and increased the risk of tumor progression and recurrence. Hypercoagulability existed in patients with ovarian cancer, and tumor associated depression could increase platelet count in plasma and increase D-dimer level. To sum up, depression can affect the level of micro inflammation in patients with ovarian cancer. In particular, depression can reduce cellular immune responses, affect the progression free survival of ovarian cancer patients, and reduce their overall survival rate.
Identification of three molecular subtypes based on immune infiltration in ovarian cancer and its prognostic value. Liu Juan,Tan Zongjian,He Jun,Jin Tingting,Han Yuanyuan,Hu Li,Song Jukun,Huang Shengwen Bioscience reports BACKGROUND:Increasing studies suggest that tumor immune infiltration is a relative factor of prognosis in ovarian cancer (OvCa). The present study explored the composition of tumor-infiltrating immune cells (TIICs) in OvCa using CIBERSORT algorithm and further assessed their values for prognosis and therapeutic strategies by molecular subtypes. METHODS:Publicly available databases including The Cancer Genome Atlas (TCGA) and GTEx were searched. Ovarian tumor samples were available from TCGA, and normal ovarian samples were obtained from the GTEx dataset. The relative proportions of immune cell profiling in OvCa and normal samples were evaluated by CIBERSORT algorithm. Association between each immune cell subtype and survival was inferred by the fractions of 22 immune cell types. "CancerSubtypes" R-package was employed to identify the three types of molecular classification and analyze the functional enrichment in each subclass. Response to immunotherapy and anticancer drug targets was predicted via TIDE algorithm and GDSC dataset. RESULTS:Substantial variation reflecting individual difference was identified between cancer and normal tissues in the immune infiltration profiles. T cells CD4 memory activated, macrophages M1 were associated with improved overall survival (OS) as evaluated by univariate Cox regression and multivariate Cox. Three subtypes were identified by ´CancerSubtypes' R-package and every sub-cluster possessed specific immune cell characterization. Meanwhile, Cluster II exhibited poor prognosis and sensitive response to immunotherapy. CONCLUSIONS:The cellular component of immune infiltration shows remarkable variation in OvCa. Profiling of immune infiltration is useful in prediction of prognosis of OvCa. The results from profiling might be considered in therapeutic modulation. 10.1042/BSR20201431
The Ovarian Cancer Tumor Immune Microenvironment (TIME) as Target for Therapy: A Focus on Innate Immunity Cells as Therapeutic Effectors. Baci Denisa,Bosi Annalisa,Gallazzi Matteo,Rizzi Manuela,Noonan Douglas M,Poggi Alessandro,Bruno Antonino,Mortara Lorenzo International journal of molecular sciences Ovarian cancer (OvCA) accounts for one of the leading causes of death from gynecologic malignancy. Despite progress in therapy improvements in OvCA, most patients develop a recurrence after first-line treatments, dependent on the tumor and non-tumor complexity/heterogeneity of the neoplasm and its surrounding tumor microenvironment (TME). The TME has gained greater attention in the design of specific therapies within the new era of immunotherapy. It is now clear that the immune contexture in OvCA, here referred as tumor immune microenvironment (TIME), acts as a crucial orchestrator of OvCA progression, thus representing a necessary target for combined therapies. Currently, several advancements of antitumor immune responses in OvCA are based on the characterization of tumor-infiltrating lymphocytes, which have been shown to correlate with a significantly improved clinical outcome. Here, we reviewed the literature on selected TIME components of OvCA, such as macrophages, neutrophils, γδ T lymphocytes, and natural killer (NK) cells; these cells can have a role in either supporting or limiting OvCA, depending on the TIME stimuli. We also reviewed and discussed the major (immune)-therapeutic approaches currently employed to target and/or potentiate macrophages, neutrophils, γδ T lymphocytes, and NK cells in the OvCA context. 10.3390/ijms21093125
Human ovarian cancer intrinsic mechanisms regulate lymphocyte activation in response to immune checkpoint blockade. Natoli Marina,Bonito Nair,Robinson James D,Ghaem-Maghami Sadaf,Mao Yumeng Cancer immunology, immunotherapy : CII Immune checkpoint blocking antibodies are currently being tested in ovarian cancer (OC) patients and have shown some responses in early clinical trials. However, it remains unclear how human OC cancer cells regulate lymphocyte activation in response to therapy. In this study, we have established and optimised an in vitro tumour-immune co-culture system (TICS), which is specifically designed to quantify the activation of multiple primary human lymphocyte subsets and human cancer cell killing in response to PD-1/L1 blockade. Human OC cell lines and treatment naïve patient ascites show differential effects on lymphocyte activation and respond differently to PD-1 blocking antibody nivolumab in TICS. Using paired OC cell lines established prior to and after chemotherapy relapse, our data reveal that the resistant cells express low levels of HLA and respond poorly to nivolumab, relative to the treatment naïve cells. In accordance, knockdown of IFNγ receptor expression compromises response to nivolumab in the treatment naïve OC cell line, while enhanced HLA expression induced by a DNA methyltransferase inhibitor promotes lymphocyte activation in TICS. Altogether, our results suggest a 'cross resistance' model, where the acquired chemotherapy resistance in cancer cells may confer resistance to immune checkpoint blockade therapy through down-regulation of antigen presentation machinery. As such, agents that can restore HLA expression may be a suitable combination partner for immunotherapy in chemotherapy-relapsed human ovarian cancer patients. 10.1007/s00262-020-02544-5
A TGF-β associated genetic score to define prognosis and platinum sensitivity in advanced epithelial ovarian cancer. Gagno Sara,Poletto Elena,Bartoletti Michele,Quartuccio Luca,Romualdi Chiara,Garziera Marica,Scalone Simona,Sorio Roberto,Dreussi Eva,Zanusso Chiara,De Mattia Elena,Roncato Rossana,Cecchin Erika,Giorda Giorgio,De Vita Salvatore,Dal Bo Michele,Puglisi Fabio,Toffoli Giuseppe Gynecologic oncology OBJECTIVE:Epithelial ovarian cancer (EOC) is usually diagnosed at advanced stages with highly variable clinical outcomes, even among patients with similar clinical characteristics and treatments. Host immune system plays a pivotal role in EOC pathogenesis and progression. Here, we assessed the clinical significance of 192 single nucleotide polymorphisms (SNPs) on 34 immune-system related genes in EOC patients. METHODS:Two hundred and thirty advanced EOC patients treated with platinum-based chemotherapy were included. Germ-line DNA was analyzed with Illumina GoldenGate Genotyping Assay. RESULTS:Nineteen polymorphisms were significantly associated with overall survival (OS), 17 with progression free survival (PFS) and 20 with platinum-free interval (PFI). Of the 8 polymorphisms associated with all three outcomes, 7 SNPs belonged to genes involved in the TGF-β pathway. A genetic score was built considering the unfavourable genotypes (UGs) of these 7 polymorphisms (group 0-2 UGs: presence of 0, 1, or 2 UGs; group 3-4 UGs: 3 or 4 UGs; group 5-7: 5, 6, or 7 UGs). According to this score, OS decreased as the number of UGs increased (median OS: 0-2 UGs = not reached, 3-4 UGs = 44.6 and 5-7 UGs = 19.3 months, p < 0.0001). The same trend was observed also for PFS (median PFS: 0-2 UGs = 21.5, 3-4 UGs = 17.3 and 5-7 UGs = 11 months, p < 0.0001) and PFI (median PFI: 0-2 UGs = 16.6, 3-4 UGs = 9.8 and 5-7 UGs = 3.8 months, p < 0.0001). The score was validated by permutation analysis. CONCLUSIONS:The proposed TGF-β pathway score could be useful to define prognosis and platinum sensitivity of advanced EOC patients. 10.1016/j.ygyno.2019.10.019
Th17-inducing autologous dendritic cell vaccination promotes antigen-specific cellular and humoral immunity in ovarian cancer patients. Block Matthew S,Dietz Allan B,Gustafson Michael P,Kalli Kimberly R,Erskine Courtney L,Youssef Bahaaeldin,Vijay Geraldine V,Allred Jacob B,Pavelko Kevin D,Strausbauch Michael A,Lin Yi,Grudem Megan E,Jatoi Aminah,Klampe Carolyn M,Wahner-Hendrickson Andrea E,Weroha S John,Glaser Gretchen E,Kumar Amanika,Langstraat Carrie L,Solseth Mary L,Deeds Michael C,Knutson Keith L,Cannon Martin J Nature communications In ovarian cancer (OC), IL-17-producing T cells (Th17s) predict improved survival, whereas regulatory T cells predict poorer survival. We previously developed a vaccine whereby patient-derived dendritic cells (DCs) are programmed to induce Th17 responses to the OC antigen folate receptor alpha (FRα). Here we report the results of a single-arm open-label phase I clinical trial designed to determine vaccine safety and tolerability (primary outcomes) and recurrence-free survival (secondary outcome). Immunogenicity is also evaluated. Recruitment is complete with a total of 19 Stage IIIC-IV OC patients in first remission after conventional therapy. DCs are generated using our Th17-inducing protocol and are pulsed with HLA class II epitopes from FRα. Mature antigen-loaded DCs are injected intradermally. All patients have completed study-related interventions. No grade 3 or higher adverse events are seen. Vaccination results in the development of Th1, Th17, and antibody responses to FRα in the majority of patients. Th1 and antibody responses are associated with prolonged recurrence-free survival. Antibody-dependent cell-mediated cytotoxic activity against FRα is also associated with prolonged RFS. Of 18 patients evaluable for efficacy, 39% (7/18) remain recurrence-free at the time of data censoring, with a median follow-up of 49.2 months. Thus, vaccination with Th17-inducing FRα-loaded DCs is safe, induces antigen-specific immunity, and is associated with prolonged remission. 10.1038/s41467-020-18962-z
Plasma Gelsolin Inhibits CD8 T-cell Function and Regulates Glutathione Production to Confer Chemoresistance in Ovarian Cancer. Asare-Werehene Meshach,Communal Laudine,Carmona Euridice,Han Youngjin,Song Yong Sang,Burger Dylan,Mes-Masson Anne-Marie,Tsang Benjamin K Cancer research Although initial treatment of ovarian cancer is successful, tumors typically relapse and become resistant to treatment. Because of poor infiltration of effector T cells, patients are mostly unresponsive to immunotherapy. Plasma gelsolin (pGSN) is transported by exosomes (small extracellular vesicle, sEV) and plays a key role in ovarian cancer chemoresistance, yet little is known about its role in immunosurveillance. Here, we report the immunomodulatory roles of sEV-pGSN in ovarian cancer chemoresistance. In chemosensitive conditions, secretion of sEV-pGSN was low, allowing for optimal CD8 T-cell function. This resulted in increased T-cell secretion of IFNγ, which reduced intracellular glutathione (GSH) production and sensitized chemosensitive cells to cis-diaminedichloroplatinum (CDDP)-induced apoptosis. In chemoresistant conditions, increased secretion of sEV-pGSN by ovarian cancer cells induced apoptosis in CD8 T cells. IFNγ secretion was therefore reduced, resulting in high GSH production and resistance to CDDP-induced death in ovarian cancer cells. These findings support our hypothesis that sEV-pGSN attenuates immunosurveillance and regulates GSH biosynthesis, a phenomenon that contributes to chemoresistance in ovarian cancer. SIGNIFICANCE: These findings provide new insight into pGSN-mediated immune cell dysfunction in ovarian cancer chemoresistance and demonstrate how this dysfunction can be exploited to enhance immunotherapy. 10.1158/0008-5472.CAN-20-0788
Immune System and DNA Repair Defects in Ovarian Cancer: Implications for Locoregional Approaches. Farolfi Alberto,Gurioli Giorgia,Fugazzola Paola,Burgio Salvatore Luca,Casanova Claudia,Ravaglia Giorgia,Altavilla Amelia,Costantini Matteo,Amadori Andrea,Framarini Massimo,Ansaloni Luca,De Giorgi Ugo International journal of molecular sciences In the last few years, substantial progress has been made in the treatment of ovarian cancer, with increased knowledge about the biology of the disease. Ovarian cancer is a neoplasm strongly linked to defects in DNA repair mechanisms, where deficiency in the homologous recombination (HR) system results in a better response of ovarian cancers to therapy, whether platinum-based chemotherapy, anthracyclines, or poly (ADP-ribose) polymerase (PARP) inhibitors. More recently, it has been demonstrated that different ovarian cancer histotypes may have different immunogenicity. Interestingly, defects in HR systems are associated more frequently with higher tumor infiltrating lymphocytes, providing a rationale for developing combination therapy with immune-modulating agents and PARP inhibitors. Again, locoregional therapies combining heat shock and chemotherapy delivery have been shown to induce an anticancer immune response in vitro. Thus, the potential for locoregional therapeutic approaches that may impact the immune system, perhaps in combination with immune-modulating agents or PARP inhibitors, needs to be further explored. With this premise, we reviewed the main biological and clinical data demonstrating a strict interplay between the immune system, DNA repair mechanisms, and intraperitoneal therapies in ovarian cancer, with a focus on potential future therapeutic implications. 10.3390/ijms20102569
Meta-analysis based gene expression profiling reveals functional genes in ovarian cancer. Zhao Lin,Li Yuhui,Zhang Zhen,Zou Jing,Li Jianfu,Wei Ran,Guo Qiang,Zhu Xiaoxiao,Chu Chu,Fu Xiaoxiao,Yue Jinbo,Li Xia Bioscience reports BACKGROUND:Ovarian cancer causes high mortality rate worldwide, and despite numerous attempts, the outcome for patients with ovarian cancer are still not well improved. Microarray-based gene expressional analysis provides with valuable information for discriminating functional genes in ovarian cancer development and progression. However, due to the differences in experimental design, the results varied significantly across individual datasets. METHODS:In the present study, the data of gene expression in ovarian cancer were downloaded from Gene Expression Omnibus (GEO) and 16 studies were included. A meta-analysis based gene expression analysis was performed to identify differentially expressed genes (DEGs). The most differentially expressed genes in our meta-analysis were selected for gene expression and gene function validation. RESULTS:A total of 972 DEGs with P-value < 0.001 were identified in ovarian cancer, including 541 up-regulated genes and 431 down-regulated genes, among which 92 additional DEGs were found as gained DEGs. Top five up- and down-regulated genes were selected for the validation of gene expression profiling. Among these genes, up-regulated CD24 molecule (CD24), SRY (sex determining region Y)-box transcription factor 17 (SOX17), WFDC2, epithelial cell adhesion molecule (EPCAM), innate immunity activator (INAVA), and down-regulated aldehyde oxidase 1 (AOX1) were revealed to be with consistent expressional patterns in clinical patient samples of ovarian cancer. Gene functional analysis demonstrated that up-regulated WFDC2 and INAVA promoted ovarian cancer cell migration, WFDC2 enhanced cell proliferation, while down-regulated AOX1 was functional in inducing cell apoptosis of ovarian cancer. CONCLUSION:Our study shed light on the molecular mechanisms underlying the development of ovarian cancer, and facilitated the understanding of novel diagnostic and therapeutic targets in ovarian cancer. 10.1042/BSR20202911
Development and validation of an immune gene-set based Prognostic signature in ovarian cancer. Shen Sipeng,Wang Guanrong,Zhang Ruyang,Zhao Yang,Yu Hao,Wei Yongyue,Chen Feng EBioMedicine BACKGROUND:Ovarian cancer (OV) is the most lethal gynecological cancer in women. We aim to develop a generalized, individualized immune prognostic signature that can stratify and predict overall survival for ovarian cancer. METHODS:The gene expression profiles of ovarian cancer tumor tissue samples were collected from 17 public cohorts, including 2777 cases totally. Single sample gene set enrichment (ssGSEA) analysis was used for the immune genes from ImmPort database to develop an immune-based prognostic score for OV (IPSOV). The signature was trained and validated in six independent datasets (n = 519, 409, 606, 634, 415, 194). FINDINGS:The IPSOV significantly stratified patients into low- and high-immune risk groups in the training set and in the 5 validation sets (HR range: 1.71 [95%CI: 1.32-2.19; P = 4.04 × 10] to 2.86 [95%CI: 1.72-4.74; P = 4.89 × 10]). Further, we compared IPSOV with nine reported ovarian cancer prognostic signatures as well as the clinical characteristics including stage, grade and debulking status. The IPSOV achieved the highest mean C-index (0.625) compared with the other signatures (0.516 to 0.602) and clinical characteristics (0.555 to 0.583). Further, we integrated IPSOV with stage, grade and debulking, which showed improved prognostic accuracy than clinical characteristics only. INTERPRETATION:The proposed clinical-immune signature is a promising biomarker for estimating overall survival in ovarian cancer. Prospective studies are needed to further validate its analytical accuracy and test the clinical utility. FUND: This work was supported by National Key Research and Development Program of China, National Natural Science Foundation of China and Natural Science Foundation of the Jiangsu Higher Education Institutions of China. 10.1016/j.ebiom.2018.12.054
Targeting tumor microenvironment in ovarian cancer: Premise and promise. Jiang Yuting,Wang Chengdi,Zhou Shengtao Biochimica et biophysica acta. Reviews on cancer Ovarian cancer is the leading cause of gynecological cancer-related mortality globally. The majority of ovarian cancer patients suffer from relapse after standard of care therapies and the clinical benefits from cancer therapies are not satisfactory owing to drug resistance. Certain novel drugs targeting the components of tumor microenvironment (TME) have been approved by US Food and Drug Administration in solid cancers. As such, the passion is rekindled to exploit the role of TME in ovarian cancer progression and metastasis for discovery of novel therapeutics for this deadly disease. In the current review, we revisit the recent mechanistic insights into the contributions of TME to the development, progression, prognosis prediction and therapeutic efficacy of ovarian cancer via modulating cancer hallmarks. We also explored potentially promising predictive and prognostic biomarkers for ovarian cancer patients. 10.1016/j.bbcan.2020.188361