Hostname: page-component-7bb8b95d7b-l4ctd Total loading time: 0 Render date: 2024-09-28T17:07:09.685Z Has data issue: false hasContentIssue false

Efficacy of immune checkpoint inhibitor monotherapy or combined with other small molecule-targeted agents in ovarian cancer

Published online by Cambridge University Press:  24 January 2023

Munawaer Muaibati
Affiliation:
Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Abasi Abuduyilimu
Affiliation:
Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Tao Zhang
Affiliation:
Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Yun Dai
Affiliation:
Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Ruyuan Li
Affiliation:
Department of Gynecology and Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
Fanwei Huang
Affiliation:
Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Kexin Li
Affiliation:
Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Qing Tong
Affiliation:
Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Xiaoyuan Huang
Affiliation:
Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
Liang Zhuang*
Affiliation:
Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Author for correspondence: Liang Zhuang, E-mail: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Ovarian cancer is the most lethal female reproductive system tumour. Despite the great advances in surgery and systemic chemotherapy over the past two decades, almost all patients in stages III and IV relapse and develop resistance to chemotherapy after first-line treatment. Ovarian cancer has an extraordinarily complex immunosuppressive tumour microenvironment in which immune checkpoints negatively regulate T cells activation and weaken antitumour immune responses by delivering immunosuppressive signals. Therefore, inhibition of immune checkpoints can break down the state of immunosuppression. Indeed, Immune checkpoint inhibitors (ICIs) have revolutionised the therapeutic landscape of many solid tumours. However, ICIs have yielded modest benefits in ovarian cancer. Therefore, a more comprehensive understanding of the mechanistic basis of the immune checkpoints is needed to improve the efficacy of ICIs in ovarian cancer. In this review, we systematically introduce the mechanisms and expression of immune checkpoints in ovarian cancer. Moreover, this review summarises recent updates regarding ICI monotherapy or combined with other small-molecule-targeted agents in ovarian cancer.

Type
Review
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

Ovarian cancer is the third most frequently diagnosed gynaecological malignancy and is the most lethal of all gynaecological malignancies worldwide, being responsible for 5% of all cancer-related deaths in women each year (Refs Reference Siegel1, Reference Torre2). Because of the lack of early clinical symptoms and screening, most patients are diagnosed with metastasis of the pelvic and peritoneal cavities (Refs Reference Torre2, Reference Hennessy3, Reference Siegel4). Although surgery and systemic chemotherapy have made great advances in the last two decades, almost all patients in stages III and IV will relapse and develop resistance to chemotherapy after first-line treatment. The 5-year survival rate of these patients is less than 25% (Refs Reference Torre2, Reference Odunsi5). Consequently, new treatment strategies and paradigms are of great need for these patients. Immune checkpoint inhibitors (ICIs) have attracted tremendous attention as promising new therapeutic targets with the recent improved understanding of the molecular basis of tumour immune microenvironment. Indeed, ICIs have revolutionised the therapeutic landscape of many solid tumours. However, there are currently no approved ICIs for ovarian cancer. Ovarian cancer is known to be an immunogenic disease in which peripheral tumour-infiltrating lymphocytes (TILs) actively recognise tumour antigens and generate tumour-specific T cells to destroy tumour cells. Unfortunately, even if large numbers of tumour-specific T cells are generated in patients by immunotherapy, these T cells fail to destroy tumour cells in vivo (Ref. Reference Anadon6). Previous studies have reported many mechanisms for this failure. For example, ovarian cancer has an extraordinarily complex immunosuppressive tumour microenvironment (TME) that is full of a large number of negative immune regulatory components, such as myeloid-derived suppressor cells (MDSCs), tumour-associated macrophages (TAMs), regulatory T cells (Tregs), cytokines, soluble factors, which have been demonstrated to be immunosuppressive functions and are associated with tumour invasiveness, spread and angiogenesis (Refs Reference Maiorano7, Reference Yin8, Reference Kolomeyevskaya9, Reference Yuan10, Reference Curiel11). Furthermore, immune checkpoint molecules have been identified as crucial regulators of the immune response. The binding of immune checkpoint receptor to ligand negatively regulates T cells activation and weakens antitumour immune responses by delivering immunosuppressive signals, ultimately leading to escape of tumour cells from immune destruction (Ref. Reference Odunsi5). ICIs could effectively prevent this effect. However, ICIs have yielded modest benefits in ovarian cancer. Therefore, a more comprehensive understanding of the mechanistic basis of the immune checkpoints is needed to improve the efficacy of ICIs in ovarian cancer.

Immune checkpoints in ovarian cancer

Immune checkpoints are a series of inhibitory regulators that directly regulate the initiation, duration and magnitude of immune responses. Normally, when the immune response is activated, immune checkpoints work as negative regulators, suppressing the immune responses, maintaining self-tolerance and preventing damage to normal tissues (Ref. Reference Edner12). However, tumour cells selectively utilise these inhibitory regulatory mechanisms to suppress effector T cells, leading to immune escape of tumour cells (Ref. Reference Jiang13) (Fig. 1). Therefore, a comprehensive understanding of the immune checkpoints in ovarian cancer is needed. We summarise immune checkpoints and functions in Table 1.

Fig. 1. Neo-antigens derived from tumour cells to CTL through MHC class I–TCRs and a co-stimulation signal of CD80 and/or CD86–CD28 interactions, CTLs are subsequently activated to destroy tumour cells. However, tumour cells often escape immune destruction through upregulation of immune checkpoint ligands, such as programmed cell death 1 ligand 1 (PD- L1), that can bind the immune checkpoint receptors programmed cell death 1 (PD-1) on the CTLs to deliver suppressing signals, finally inhibit the proliferation and activation of CTLs. Another negative-regulate immune checkpoint molecule cytotoxic T lymphocyte protein 4 (CTLA-4) that binds CD80 and CD86 and prevents their interaction with CD28, inhibit the co-stimulation signal of CD80 and/or CD86-CD28 interactions, thus inhibit the proliferation and activation of CTLs. ICIs could effectively prevent this effect. ICIs highly specifically bind to immune checkpoints, blocking this inhibitory mechanism and thereby reactivating the anti-tumour immune response.

Table 1. Summary of immune checkpoints and functions

PD-1

The immune checkpoint molecule programmed cell death protein 1 (PD-1) is expressed on activated T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, dendritic cells (DCs) and macrophages and interacts with two ligands, programmed death ligand 1 (PD-L1) and programmed death ligand 2 (PD-L2), to exert inhibitory effects on both peripheral lymphocytes and the TME (Refs Reference Ishida32, Reference Nishimura33). PD-1 is a type I transmembrane receptor, the cytoplasmic tail is composed of two tyrosyl residues and the N-terminal tyrosine residues constitute an immunoreceptor tyrosine-based inhibitory motif (ITIM). The C-terminal domain constitutes an immunoreceptor tyrosine-based switch motif (ITSM). When PD-1 interacts with PD-L1/PD-L2, ITIM and ITSM are phosphorylated, recruiting and activating the Src homology (SH) domains of SH-containing phosphatase (SHP), which dephosphorylates the crucial downstream intracellular signalling pathway PI3K-Akt. Ultimately, this leads to a reduction of both cytokine production and T-cell proliferation, thereby suppressing T-cell-mediated immune response (Refs Reference Ishida32, Reference Nishimura33, Reference Marasco34). In addition, when PD-1 interacts with PD-L1/L2, it upregulates the E3 ubiquitin ligases casitas B-lineage lymphoma-b (CBL-b) and c-casitas B-lineage lymphoma (c-CBL) and triggers PD-1 pathway-mediated suppression of antitumour immune responses. Tregs are strongly associated with advanced stages of ovarian cancer and have immunosuppressive effects on tumours. Terme et al. claimed that PD-1 binding to ligands could regulate the differentiation of Tregs and maintain their immunosuppressive functions. The latest study illustrated that the immunosuppressive cytokine interleukin-18 produced by tumour cells upregulates PD-1 expression on activated mature NK cells, thereby inhibiting NK cell-dependent immunosurveillance in many tumours (Ref. Reference Terme35). In addition, B cell receptor (BCR) induces the expression of PD-1 on the surface of B cells, which inhibits B cells function in tumours (Ref. Reference Okazaki15). These results indicate that PD-1 inhibits the antitumour immune response through multiple pathways and that targeted PD-1 therapies play an antitumour role in part by inhibiting Treg cells and restoring B-cell and NK-cell functions.

In ovarian cancer, Matsuzaki et al. elaborated that compared with peripheral blood lymphocytes, tumour-derived NY-ESO-1-specific CD8(+) T cells enriched co-expression of inhibitory molecules lymphocyte activation gene 3 (LAG-3) and PD-1, dual blockade of LAG-3 and PD-1 during T-cell priming efficiently augmented proliferation and cytokine production by NY-ESO-1-specific CD8 (+) T cells (Ref. Reference Matsuzaki36). Tu et al. used Oncomine and PrognoScan database analyses to investigate the expression levels and prognostic values of PD-1 in ovarian cancer, and found that the expression of PD-1 was closely associated with relatively poor survival in an advanced stage of ovarian cancer (Ref. Reference Tu37). Moreover, Rådestad et al. reported that CD8+ T cells coexpressed the immune checkpoints LAG-3, PD-1 and T-cell immunoglobulin domain and mucin domain-3 (TIM-3) in tumours, the most common combination being PD-1 and TIM-3, and dual blockade of these molecules improved CD8+ T-cell response to non-specific stimulate in the TME by synthesising effector (Ref. Reference Rådestad38). Another study has shown that CD8+ T cells that do not express LAG-3, PD-1 and TIM-3 are beneficial for OS (Ref. Reference Hensler39). These results provide new insights to investigate the simultaneous blockade of multiple immune checkpoints in the treatment of ovarian cancer.

On the contrary, PD-L1 has received a great deal of attention. It has been reported that PD-L1 is not expressed in normal tissue but is increased in ovarian cancer, PD-L1 expression is significantly higher in malignant disease than in benign/borderline disease, tumour cells lysis by cytotoxic T lymphocytes (CTLs) was attenuated when PD-L1 was overexpressed and promoted when it was silenced in mouse ovarian cancer cells, and PD-L1 expression in tumour cells promotes peritoneal dissemination by repressing CTL function (Refs Reference Hensler39, Reference Maine40, Reference Abiko41, Reference Parvathareddy42). In addition, PD-L1 expression in tumours correlates with FIGO stage of ovarian cancer. Obviously, the PD-1/PD-L1 signalling pathway plays a crucial role in the occurrence and development of ovarian cancer. However, some studies have reported different results, with no significant correlation between PD-1 expression and infiltration of effector T cells in tumours (Refs Reference Hensler39, Reference Darb-Esfahani43). In conclusion, the relationship between PD-1 and PD-L1 expression and the prognosis of ovarian cancer patients remains controversial, and more studies are needed to investigate the role of immune checkpoints in ovarian cancer.

CTLA-4

Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), also known as CD152, is a leucocyte differentiation antigen and a transmembrane receptor. Its intracellular structural domain consists of 36 amino acids forming the ITIM, which plays an opposite role in the intracellular ITAM structural stimulatory molecule CD28 (Ref. Reference Lingel17). CTLA-4 is mainly expressed on the surface of activated CD4+ and CD8+ effector T cells and on Tregs, and is involved in early T-cell activation in secondary lymphoid organs. In ovarian cancer, CTLA-4 competitively binds to the same ligands as the costimulatory receptor CD28, namely, B7 (B7-1: other name CD80; B7-2: other name CD86), expressed on the surface of antigen-presenting cells (APCs) but with greater affinity (Refs Reference Egen16, Reference Linsley44). CTLA-4 interacts with ligands to upregulate inhibitory signals, inhibiting the cell cycle progression of T cells from the G1 to S phase and attenuating or even terminating T-cell immune response (Ref. Reference Krummel45). Furthermore, another biological function of CTLA-4 is endocytosis, which induces CD80/CD86 endocytosis and downregulates CD80 and CD86 expression, thereby further inhibiting T-cell function.

Recently, an increasing number of experiments have demonstrated the important roles of CTLA-4 in ovarian cancer. Jaikumar et al. reported that one-third to half of CD8+ TILs coexpressed PD-1 and CTLA-4 in ovarian cancer, and PD-1 + CTLA-4 + CD8+ TILs have more severe dysfunctional features than PD-1+ or CTLA-4+ TILs. Dual blockade of PD-1 and CTLA-4 reverses CD8+ TIL dysfunction and activates antitumour immune responses in the majority of mice (Ref. Reference Duraiswamy46). Furthermore, some experiments have provided evidence that TILs usually express multiple immune checkpoints in patients with ovarian cancer. In total, CTLA-4 plays a crucial role in immune escape, and dual blockade may be an effective strategy to activate antigen-specific effector T cells.

LAG-3

Lymphocyte activation gene 3 (LAG-3) is mainly expressed on the surface of activated T cells. Its extracellular molecular structure is similar to that of CD4, and it interacts stably with major histocompatibility complex (MHC)-II molecules in a non-competitive manner with a significantly higher affinity than CD4 (Ref. Reference Huard47). This interaction suppresses T-cell receptor (TCR)-mediated T-cell proliferation and activation by downregulation of intracellular STAT5 phosphorylation and reducing CD3 and TCR expression (Refs Reference Duraiswamy46, Reference Grosso48). However, several studies reported that the binding of LAG-3 to MHC-II was not the main inhibitory mechanism. Galectin-3 is the second major LAG-3 functional ligand independent of MHC-II. Interaction of LAG-3 with galectin-3 is needed for galectin-3-mediated CD8(+) T cells inhibition in vitro (Ref. Reference Kouo49). LSECtin, a cell surface lectin constitutively expressed in many tumour cells, is also a ligand for LAG-3, and blocking the interaction of LAG-3 with LSECtin restores interferon-γ secretion and regulates the function of CD8+ T cells and NK cells (Ref. Reference Xu50). Notably, in 2019, Jun et al. reported another ligand, FGL1, is produced by human cancer cells and that blocking the FGL1–LAG-3 interaction preferentially stimulates T cells in tumours and can treat established mouse tumours (Ref. Reference Wang51). Furthermore, LAG-3 negatively regulates T-cell proliferation by upregulating Tregs, thereby promoting the immune escape of tumour cells (Ref. Reference Workman19). In ovarian cancer, researchers observed that CD8+ T cells coexpressing LAG-3 (+) PD-1 (+) have more severe dysfunction than LAG-3 (+) PD-1 (−) or LAG-3 (−) PD-1 (−) subsets. Dual blockade of LAG-3 and PD-1 significantly increased effector T-cell function and dual anti-LAG-3/anti-PD-1 antibody treatment cured most mice of established tumours that were largely resistant to single-antibody treatment. But not by blocking either molecule alone (Refs Reference Matsuzaki36, Reference Woo52, Reference Huang53). This suggests that dual blockade of these molecules remains one of the most promising regimens to explore immunotherapy.

TIM-3

T-cell immunoglobulin domain and mucin domain-3 (TIM-3) are expressed on T cells, Th1 cells, cytotoxic T cells, Treg cells and innate immune cells (Ref. Reference Das54). A study reported that the interaction of TIM-3 and galectin-9 secreted by tumour cells increased apoptosis of CD8+ TIL cells in colorectal cancer (Refs Reference Kang22, Reference Zhu55). TIM-3 interacts with ligands ceacam1 to inhibit the function of effector T cells, and ligands galectin-9 and ceacam1 have a synergistic effect on the TIM-3 signalling pathway. High-mobility group box1 (HMGB1) is another ligand of TIM-3. TIM-3 interacts with HMGB1 to suppress nucleic acid-mediated innate immune responses (Ref. Reference Chiba21). PtdSer (PS) is a non-protein ligand for TIM-3, binds to TIM-3 and recognises apoptotic cells, resulting in clearance of apoptotic cells by resident phagocytes (Ref. Reference Santiago56).

In ovarian cancer, TIM-3 is involved in tumourigenesis and progression by suppressing immunity. In 2017, Xu et al. observed that TIM-3+ CD4T cells isolated from tumour tissue were significantly higher than those isolated from normal tissue. This phenomenon was related to tumour grade, with further increased expression of TIM-3 in T cells (CD4+ and CD8+) from high-grade tumours compared with other lower-grade tumours (Ref. Reference Xu57). Another study showed that TIM-3 + Foxp3 + CD4T cells induce TIM-3 + Tregs in ovarian cancer. It is well known that TIM-3 + FoxP3 + Treg is a promoter of T-cell dysfunction in tumours, thus TIM-3-FoxP3 + Tregs may cause strong immunosuppression in ovarian cancer (Refs Reference Yan58, Reference Sakuishi59). Overall, TIM-3 may negatively regulate antitumour immune responses through various T-cell subsets.

TIGIT

T-cell immunoglobulin and ITIM domain (TIGIT) is a promising new target for cancer immunotherapy. Similar to CD28 and CTLA-4, it competes with CD226 for the same ligands CD155 (PVR) and CD112 (PVRL2, nectin-2), thereby inhibiting the function of T cells and NK cells. TIGIT is upregulated by immune cells in tumours, and its ligands CD155 and CD112 are overexpressed in various tumour cells, but almost absent in normal cells. Whelan et al. reported that ovarian cancers having the highest percentage of PVRPVRL2+ tumour cells, and a combination of PVRIG blockade with TIGIT or PD-1 blockade further increased T-cell activation (Ref. Reference Whelan60). Furthermore, TIGIT was observed to suppress the antitumour immune response by enhancing the CD4 + Treg cell response (Refs Reference Li61, Reference Harjunpää62, Reference Maas63). Chen et al. reported that anti-TIGIT treatment reduced CD4 + Tregs without affecting CD4+ or CD8+ T cells or NK cells (Ref. Reference Chen64).

BTLA

B and T lymphocyte attenuator (BTLA) is the third coinhibitory molecule observed from the CD28 family and is structurally similar to CTLA-4 and PD-1. BTLA is constitutively expressed on resting T cells and upon activation continues to be expressed. Herpesvirus entry mediator (HVEM), a ligand for BTLA, binds to BTLA, induces its phosphorylation and binds to the tyrosine phosphatase SHP-2, suppressing the immune responses and leading to tumour immune tolerance (Refs Reference Han27, Reference Yu65).

An early study elucidated that BTLA is overexpressed on the surface of CD4+ and CD8+ T cells in patients with various tumours. In addition, HVEM expression was elevated in ovarian cancer cells compared with benign tissues, and T cells numbers and secretion of anti-tumour cytokines were increased in HVEM (−) ovarian cancer (Ref. Reference Zhang66). Chen et al. reported that BTLA is mainly expressed in B lymphocytes and its detection in cancer tissues predicts poor outcome of epithelial ovarian cancer (EOC) patients. Preclinical experiments showed that blocking BTLA in combination with chemotherapy significantly reduced peritoneal tumour volume in tumour-bearing mice (Ref. Reference Chen67).

VISTA

V-domain immunoglobulin suppressor of T-cell activation (VISTA) is a recently discovered negative regulator that is persistently expressed on naive T cells. VISTA acts as a ligand, receptor or both in the TME. VISTA is overexpressed in a variety of tumours, and blocking the VISTA signalling pathway can enhance the antitumour immune response in mice (Refs Reference Lines68, Reference ElTanbouly69). Some studies found that VISTA was almost absent in normal ovarian epithelial cells but was overexpressed in ovarian cancer, and anti-VISTA therapy markedly prolonged the survival of mice bearing tumours that expressed high VISTA (Ref. Reference Mulati70). Notably, Zong et al. gave the opposite conclusion that VISTA expression has been associated with favourable clinical outcomes in patients with high-grade serous ovarian cancer (Ref. Reference Zong71). The differences in these results were related to the variability of the study samples, the complex immunosuppressive microenvironment of ovarian cancer and the weak immunosuppressive functions of VISTA. Targeting VISTA might be a way to enhance antitumour immune responses. However, more studies are still needed to investigate the effects of NISTA on humans.

IDO-1

Tryptophan is involved in multiple catabolic processes and is required for T-cell activation. Indoleamine 2,3-dioxygenases (IDO-1) are tryptophan-degrading enzymes that degrade tryptophan to kynurenine, and the depletion of tryptophan and generation of kynurenine play important immunosuppressive functions by activating Tregs and MDSCs, suppressing the functions of effector T and NK cells. Moreover, IDO-1 modulates the downstream effector pathway, promoting neovascularisation of solid tumours, which promotes tumour growth (Refs Reference Liu72, Reference Wainwright73, Reference Mellor74). Several studies claimed that upregulation of IDO-1 expression in ovarian cancer suppresses T-cell expansion and reduces the number of CD8 + TILs, resulting in a poor prognosis in patients. Another study described that IDO-1 induced PD-1 expression in T-cell ovarian cancer, which was positively associated with paclitaxel resistance (Refs Reference Qian75, Reference Okamoto76, Reference Takao77, Reference Amobi-McCloud78). Importantly, these results demonstrate that IDO-1 plays an important role in ovarian cancer.

Clinical trials of ICI monotherapy in ovarian cancer

In the second part, we describe the mechanisms of immune checkpoints and their crucial roles in ovarian cancer. Therefore, we believe that immune checkpoints are the most promising targets for ovarian cancer therapy. ICIs are small-molecule agents that target immune checkpoints and specifically recognise and bind immune checkpoint molecules, reducing immunesuppression and thus enhancing antitumour immune responses. Currently, several ICIs have been approved by the U.S. Food and Drug Administration (FDA) for cancer treatment (Refs Reference Boland79, Reference Wolchok80, Reference Hodi81). Although none have been approved for ovarian cancer, several clinical trials are currently underway to evaluate the clinical activity of ICIs in ovarian cancer. Table 2 lists the completed clinical trials of ICI monotherapy in ovarian cancer.

Table 2. Clinical trials of ICI monotherapy in ovarian cancer

PD-1 inhibitors

Nivolumab

Nivolumab is an immunoglobulin G4 (IgG4) monoclonal antibody that targets PD-1 receptors. Nivolumab binds to PD-1 block negative/suppressing signal delivery to the T cell, activating them and enhancing host anti-tumour immunity. To date, nivolumab has been approved by the FDA for treatment of various tumours.

In 2014, Hamanishi et al. evaluated the efficacy of nivolumab in ovarian cancer in a phase II clinical trial that enrolled 20 patients with platinum-resistant ovarian cancer. In this trial, patients were sequentially assigned to the high-dose group (nivolumab 3 mg/kg; n = 10) and the low-dose group (nivolumab 1 mg/kg; n = 10) and received nivolumab monotherapy. A better objective response ratio (ORR) [20%; 95% confidence interval (CI), 2.5–55.6] was found in the 3 mg/kg cohort compared with an ORR of 10% (95% CI, 0.3–44.5) in the 1 mg/kg cohort. The incidence of treatment-related adverse events (TRAEs) did not differ between the two groups. Consequently, it can be concluded that ovarian cancer patients may benefit more from nivolumab 3 mg/kg (Ref. Reference Hamanishi83). Subsequently, in 2021, Hamanishi et al. compared the efficacy of nivolumab alone or chemotherapy in patients with platinum-resistant ovarian cancer, demonstrating that the nivolumab group was more well tolerated than the chemotherapy group. However, no remarkable clinical benefit was observed in this study, with a median overall survival (OS) of 10.1 months (95% CI, 8.3–14.1) in the nivolumab group and 12.1 months (95% CI, 9.3–15.3) in the chemotherapy group, and there was no significant difference in OS between groups (Ref. Reference Hamanishi84). Normann et al. also evaluated the efficacy of nivolumab in patients with platinum-resistant ovarian cancer and reported that the disease control rate (DCR) was 44% (95% CI, 19–87), the median OS was 30 weeks (95% CI, 14–42) and the progression free survival (PFS) was 15 weeks (95% CI, 13–17) (Ref. Reference Normann101). This study reported similar DCR and PFS to the trial reported by Hamanishi in 2014.

In conclusion, nivolumab monotherapy has limited clinical efficacy in patients with advanced or platinum-resistant ovarian cancer. However, nivolumab showed an acceptable safety profile, suggesting that more investigations would be valuable to elucidate the clinical efficacy of nivolumab in ovarian cancer.

Pembrolizumab

Pembrolizumab (Keytruda) is a humanised monoclonal IgG4 antibody that blocks the PD-1 pathway and has been extensively investigated in a variety of malignancies.

In a phase Ib trial (NCT02054806), the clinical efficacy and safety of pembrolizumab monotherapy in patients with PD-L1-expressing advanced ovarian cancer was evaluated, TRAEs occurred in 19 (73.1%) patients. One grade 3 TRAEs (increased plasma transaminase level) occurred. No deaths and no treatment discontinuations because of TRAEs occurred. The ORR was 11.5%, with seven patients (26.9%) having stable disease, and the median PFS and OS were 1.9 (95% CI, 1.8–3.5) and 13.8 (95% CI, 6.7–18.8) months, respectively (Ref. Reference Varga88). Matulonis et al. reported a phase II clinical trial of 376 patients with advanced recurrent ovarian cancer in which pembrolizumab monotherapy; ORR was 8%, DCR was 37% and PFS was 2.1 months. The study also found that high levels of PD-L1 were related to an increased clinical efficacy of pembrolizumab. Nishio et al. also reported in advanced ovarian cancer pembrolizumab monotherapy had an ORR of 19.0% (95% CI, 5.4–41.9) and seemed to increase with increasing PD-L1 expression. A total of 13 (61.9%) patients had TRAEs, and five (23.8%) had grade 3–4 TRAEs (Refs Reference Matulonis85, Reference Nishio89). These results indicated that the levels of PD-L1 expression in tumour cells may be a valid predictor of disease prognosis. However, a variety of trials have also reported that patients can benefit from the combination of ICIs with other therapies regardless of their PD-L1 expression levels.

Overall, these trials demonstrated an ORR of 5–20% with manageable toxicities in patients with advanced ovarian cancer with pembrolizumab monotherapy. Further studies are needed to identify appropriate predictors to facilitate pembrolizumab efficacy.

Dostarlimab

Dostarlimab (TSR-042) is another anti-PD-1 inhibitor that interacts with the PD-1 receptor with high affinity. Currently, no clinical trials have reported the clinical activity of dostarlimab in ovarian cancer. However, multiple phase III clinical trials (NCT04679064; NCT03602859; NCT03806049) are being performed to test dostarlimab as a monotherapy or in combination with other agents in ovarian cancer, and we expect promising outcomes from these treatments.

PD-L1 inhibitors

Atezolizumab

Atezolizumab is a fully humanised monoclonal antibody that selectively targets PD-L1 to prevent interaction with PD-1. Liu et al. investigated single-agent atezolizumab in 12 patients with recurrent EOC in a first-in-human phase 1 study. Antitumour activity of atezolizumab was observed in two patients with mostly grade ≤ 2 TRAEs and no grade ≥ 4 TRAEs were reported (Ref. Reference Liu95). Overall, atezolizumab monotherapy was generally well tolerated in patients with recurrent EOC, and its clinical efficacy warrants further investigation.

Avelumab

Avelumab is a fully humanised IgG1 monoclonal antibody against PD-L1. It is the only agent that kills cancer cells using both antibody-dependent cell-mediated cytotoxicity and immune checkpoint inhibition simultaneously. Disis et al. first investigated the clinical activity of ipilimumab in previously treated patients with recurrent or refractory ovarian cancer in a phase Ib trial (NCT01772004). The ORR was 9.6% (95% CI, 5.1–16.2), the 1-year PFS rate was 10.2% (95% CI, 5.4–16.7) and the median OS was 11.2 months (95% CI, 8.7–15.4 months). Other frequent TRAEs were fatigue (17 [13.6%]), diarrhoea (15 [12.0%]) and nausea (14 [11.2%]). Grade 3 or higher TRAEs occurred in nine patients (7.2%). Twenty-one patients (16.8%) had TRAEs of any grade (Ref. Reference Disis99). Another phase 3 trial evaluated avelumab alone or avelumab plus chemotherapy compared with chemotherapy alone in patients with platinum-resistant or platinum-refractory ovarian cancer. The results showed that the median OS was 15.7 months (95% CI, 12.7–18.7), 13.1 months (11.8–15.5) and 11.8 months (8.9–14.1) in the combination group, chemotherapy group and avelumab group, respectively. Here, we found no significant clinical benefit of avelumab alone or in combination with chemotherapy compared with chemotherapy, and worse PFS and OS were observed in the avelumab group (Ref. Reference Pujade-Lauraine96). Overall, avelumab showed limited clinical activity in recurrent or refractory ovarian cancer. Monk et al. compared chemotherapy plus avelumab, chemotherapy followed by avelumab maintenance and chemotherapy alone in stage III–IV epithelial ovarian, fallopian tube or peritoneal cancer. The median PFS was 18⋅1 months [14.8 to not estimable (NE)] with avelumab combination treatment and 16.8 months (95% CI, 13⋅5 to not estimable) with avelumab maintenance. No significant clinical benefit was observed with either avelumab maintenance therapy or chemotherapy plus avelumab in this trial. More studies are needed to evaluate the clinical efficacy of avelumab as a first-line therapy (Ref. Reference Monk98).

CTLA-4 inhibitors

Ipilimumab is the first FDA-approved ICI that effectively blocks the CTLA-4 pathway. A phase II study (NCT01611558) in 40 patients evaluated the safety and efficiency of ipilimumab monotherapy in recurrent platinum-sensitive ovarian cancer. This trial found that the incidence of grade ≥3 TRAEs was 50%, and the best ORR was 15% (5.7–29.8). However, studies on ipilimumab are limited. Randomised phase 2 or 3 clinical trials of CTLA-4-targeted agents in ovarian cancer patients have not reported a clear overall survival benefit, either alone or in combination with available agents.

In conclusion, ICIs have yielded only modest responses as monotherapy for advanced or recurrent ovarian cancer. There are extensive challenges for further clinical applications of ICIs alone in ovarian cancer, such as the limited effectiveness of monotherapy, the lack of investigation of identified biomarkers to predict prognosis and serious TRAEs and primary and acquired resistance. Therefore, further research is needed to develop combination approaches to allow more patients to benefit from ICIs.

Clinical trials of ICIs combined with other small-molecule-targeted agents in ovarian cancer

The antitumour function of ICIs depends on TILs, and the activation of TILs requires immunogenic tumour-specific antigens (Refs Reference Linette102, Reference Cheng103). The standard treatment for ovarian cancer includes surgery followed by platinum-based chemotherapy. Chemotherapy can lead to the destruction of cancer cells and the release of immunogenic molecules (Ref. Reference Bezu104). Combining gemcitabine chemotherapy drugs with a CTLA-4 blockade could induce a potent CD4+ and CD8+ T-cell-dependent antitumour immune response. Furthermore, small-molecule-targeted agents induce tumour cell death and lead to the release of large amounts of neoantigens, thereby enhancing the infiltration and activation of effector T cells in the TME. Therefore, the combination of ICIs with other small-molecule-targeted agents achieves cumulative or synergistic therapy to show the greatest antitumour immune responses. Clinical trials of ICIs combined with other small-molecule-targeted agents in ovarian cancer are listed in Table 3.

Table 3. Clinical trials of ICIs combined with other small-molecule-targeted agents in ovarian cancer

Multiple ICIs in combination

Several preclinical studies have demonstrated that 33–50% of CD8 (+) TILs coexpress more than one immune checkpoints, and that blocking any of these immune checkpoints compensatively increase the expression of other immune checkpoints, and that tumour cells may choose this replacement immunosuppressive molecule to continue evading attack from the immune system (Refs Reference Rådestad38, Reference Li123). Tumour cells also overexpress several immune checkpoints ligands, to synergistically exploit coexpression of immune checkpoints at the T-cell surface as an immune escape mechanism (Ref. Reference Curdy124). Huang et al. observed that dual blockade of PD-1 with CTLA-4 or LAG-3 synergistically enhanced effector T-cell function and led to tumour rejection in mouse ovarian tumours (Ref. Reference Huang125). Therefore, blocking multiple immune checkpoints could improve the efficacy of ICIs in ovarian cancer.

A phase 1, 2 trial NCT03287674 evaluated ipilimumab and nivolumab in metastatic ovarian cancer and showed that the ORR was 16.7%, and the stable disease rate was 83.3%. Although it had limited efficacy, the combination strategy showed better efficacy than monotherapy. Subsequently, Zamarin et al. reported results from the phase II trial NCT02498600, which evaluated ipilimumab plus nivolumab compared with nivolumab alone in patients with persistent or recurrent EOC. Grade ≥3 TRAEs occurred in 33 and 49% of patients in the nivolumab and combination groups, respectively. Within 6 months of treatment responses occurred in six (12.2%) patients in the nivolumab group and 16 (31.4%) patients in the nivolumab plus ipilimumab group (OR, 3.28; 85% CI, 1.54 to infinity), and the median PFS was 2 and 3.9 months in the nivolumab and combination groups, respectively. These results demonstrated that combination therapy for persistent or recurrent EOC improved response rates and prolonged PFS compared with nivolumab alone (Ref. Reference Zamarin82). Overall, the combination regimens showed superior efficacy in patients with persistent or recurrent EOC.

Combination of ICIs and PARP inhibitors

In tumours, the rapid expansion of cancer cells is prone to DNA damage, which requires rapid DNA damage repair (DDR). There are two most common modalities of DDR: DNA single-strand break (SSB) repair involving poly (ADP-ribose) polymerase (PARP) enzymes and homologous recombination repair in which BRCA1/2 plays an important role (Ref. Reference Venkitaraman126). During DNA replication, when SSB repair is blocked, the replication fork collides with the unrepaired SSBs, forming a double-strand break (DSB). Therefore, if only the SSB repair pathway is blocked, cells can still rely on HR to repair DSBs. However, in HR deficiency (HRD) cancer cells, PARPi impairs the repair of DNA SSBs, rendering DSBs ineffective and leading to the accumulation of damage, chromosomal rearrangements, genomic instability and synthetic lethality (Refs Reference Venkitaraman126, Reference Konstantinopoulos127, Reference Pilié128), which means, patients with BRCA1/2 mutations are particularly sensitive to PARPi (Refs Reference Foo129, Reference Zheng130). It has been reported that HRD-related DDR was observed in approximately 40–50% of patients with ovarian cancer (Ref. Reference da Cunha Colombo Bonadio131), and the majority of them are strongly associated with BRCA1/2 mutation. BRCA1/2 mutation EOC showed higher neoantigen load and PD-L1 expression compared with BRCA1/2 wild type and HR proficient. Ding et al. described the results from preclinical studies; PARPi drives powerful local and systemic antitumour immunity in mice bearing BRCA1-deficient ovarian tumours, and this antitumour effect is further enhanced when PARPi is combined with a PD-1 inhibitor (Refs Reference Ding132, Reference Wang133, Reference Shen134). Therefore, these results provide a powerful molecular basis for PARPi in combination with ICIs playing a synergic role in ovarian cancer.

A phase 2 clinical trial NCT02484404 enrolled 35 ovarian cancer patients and evaluated the efficacy of olaparib and durvalumab; the ORR was 14% (5/35; 95% CI, 4.8–30.3), and the DCR was 71% (25/35; 95% CI, 53.7–85.4) (Ref. Reference Lampert112). In addition, Konstantinopoulos et al. reported results from an open-label, single-arm, phase 1 and 2 trial that evaluated niraparib in combination with pembrolizumab in patients with recurrent ovarian carcinoma. The ORR was 18% (90% CI, 11–29), and the DCR was 65% (90% CI, 54–75). Overall, these results suggested that niraparib combined with pembrolizumab has a favourable safety profile tolerable and showed promising antitumour activity in patients with recurrent ovarian cancer, and this combination strategy may represent a new choice for these individuals (Ref. Reference Konstantinopoulos135). In addition, several ongoing trials (NCT02657889, NCT04169841) are also investigating the efficacy of ICIs in combination with PARPi therapy in patients with solid tumours and expect promising results.

Combination of ICIs and VEGF/VEGFR inhibitors

Vascular endothelial growth factor (VEGF) is a highly biologically active glycoprotein, and its ligand vascular endothelial growth factor receptor (VEGFR) is expressed on endothelial cells, thereby triggering angiogenesis signals. Neovascularisation in the TME plays a key role in tumour progression, invasion and metastasis. Recently, several studies have found that VEGF/VEGFR expression is significantly higher in tumour vascular cells than in normal vascular cells, and the highest levels of VEGF were observed in patients diagnosed with advanced tumours (Refs Reference Bekes136, Reference Nagy137, Reference Luo138, Reference Mahner139). VEGF inhibitors exert antitumour effects by targeting the blocking of VEGF signalling pathways, inhibiting tumour neovascularisation and causing tumour vascular regression. Currently, VEGF inhibitors have shown therapeutic efficacy in an increasing number of human cancers (Refs Reference Azam140, Reference Bergers141). Moreover, some researchers reported that PD-L1 inhibitors plus antiangiogenic agents can inhibit angiogenesis and tumour progression induced by the direct interaction of PD-L1 and VEGFR2, and this combination therapy can also overcome single-drug resistance (Ref. Reference Yang142). Furthermore, antiangiogenic therapy attempts to normalise the tumour vasculature and improve the efficiency of anticancer drug delivery, and better efficacy can be achieved with a lower dose of ICIs, which can decrease TRAEs. Combining PD-L1 antiangiogenic agents may be a potential therapeutic strategy for ovarian cancer patients.

A single-arm, phase 2 trial evaluated the combination of nivolumab and bevacizumab in 38 patients (18 had platinum-resistant and 20 had platinum-sensitive disease) with relapsed EOC. In this trial, the ORR was 40.0% (19.1–64.0%) in platinum-sensitive participants and 16.7% (95% CI, 3.6–41.4) in platinum-resistant participants. Nivolumab combined with bevacizumab has significant clinical activity in relapsed ovarian cancer patients, with greater activity in the platinum-sensitive setting (Ref. Reference Liu106). Moroney also reported similar safety and clinical activity in a platinum-resistant ovarian cancer setting, with an ORR of 15% and stable disease in eight patients (40%) (Ref. Reference Moroney109). In conclusion, ICIs combined with VEGF inhibitors have a better benefit in platinum-sensitive ovarian cancer, but their clinical efficacy in platinum-resistant ovarian cancer is still limited.

Combination of ICIs and DNMT inhibitors

In normal cells, DNA methylation is crucial for regulating gene expression and is required for the maintenance of genome stability. Aberrant DNA methylation leads to alterations in chromatin structure and silencing of tumour suppressor genes, ultimately leading to tumourigenesis (Ref. Reference Klose143).

DNA methyltransferase (DNMT) is an important epigenetic molecule for DNA methylation that can catalyse DNA methylation and inhibit gene transcription. Therefore, DNMT inhibitors can block abnormal DNA methylation during tumourigenesis, promoting the activation of tumour suppressor genes and achieving antitumour effects. Some studies have elaborated that DNMT is expressed at levels three times higher in ovarian cancer cells than in normal ovarian epithelial cells (Ref. Reference Ahluwalia144).

Guadecitabine is a second-generation hypomethylating agent, phase I/II randomised trial (NCT02901899) evaluating the clinical efficacy of guadecitabine and pembrolizumab in recurrent ovarian, peritoneal and fallopian tube carcinomas. Final results have not yet been available.

Combination of ICIs and IDO-1 inhibitors

The immunosuppressive enzyme IDO-1 catalyses the cleavage of l-tryptophan to produce a series of kynurenine metabolites that inhibit the action of CD8+ T lymphocytes (Refs Reference Stone145, Reference Li146). IDO-1 inhibitors have been reported to be synergistic with ICIs and may increase the effectiveness of ICIs in cancer patients. Several studies are evaluating the efficacy and safety of IDO-1 inhibitors plus ICIs in ovarian cancer.

Combination of ICIs and FRα inhibitors

Folate is an important regulator of cell growth and survival. The binding of folate to folate receptor α (FRα) is one of the main methods by which folate enters cells. Several studies found that FRα was selectively overexpressed in ovarian cancers, whereas expression was not detectable in normal ovarian surface epithelium (Refs Reference Choi147, Reference Scaranti148, Reference Figini149). Furthermore, FRα can be transferred into the nucleus, where it acts as a transcription factor to regulate the expression of key developmental genes in tumour cells. At the same time, folate regulates tumour growth and development by participating in a variety of intracellular signalling pathways and downregulating cell adhesion molecules (Ref. Reference Nawaz150).

A phase II trial of durvalumab in combination with the multiepitope FRα vaccine TPIV200 in 27 ovarian cancer patients observed a treatment-related grade 3 toxicity rate of 18.5%. Although the ORR in this trial was only 3.7%, all patients had increased T-cell responses at 6 weeks with a median OS of 21 months (13.5 to infinity). These observations demonstrated that a combination therapeutic strategy of ICIs and FRα inhibitors or FRα vaccines is extremely valuable (Ref. Reference Zamarin119).

Combination of ICIs and HDAC inhibitors

Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are involved in transcription, cell cycle regulation and cell transformation. Normally, histones are in a dynamic balance between acetylation and deacetylation and are coregulated by HAT and HDAC. In human cells, increased levels of histone deacetylation result in alterations of the normal cell cycle and metabolic behaviour, which ultimately induce tumours (Ref. Reference Wade151). Anti-HDAC therapy can activate histone acetylation, promote the expression of antitumour transcription factors and inhibit tumourigenesis. Furthermore, HDAC inhibitors promote the degradation of the proto-oncoprotein (Ref. Reference Eckschlager152). Several studies have observed that HDACs are frequently overexpressed in ovarian cancer and are often associated with poor prognosis (Refs Reference Li153, Reference Weichert154, Reference Yano155). ICIs combined with HDAC inhibitors may be a new treatment strategy for ovarian cancer patients. The phase II trial NCT02915523 is testing in combination with entinostat and avelumab in ovarian, fallopian tube, or primary peritoneal cancer, and we expect good outcomes from this treatment.

Combination of ICIs and RAF/MEK/ERK pathway inhibitors

Ras has been identified as an oncogene, and the RAF–MEK–ERK pathway is an important downstream effector of Ras. Ras mutation activates the RAF–MEK–ERK signalling pathway, which plays a key role in cancer cell expansion, invasion and metastasis. Hence, each component of the RAS/RAF/MEK/ERK pathway has become an important target of antitumour therapy (Refs Reference Roberts156, Reference De Luca157). Currently, RAF and MEK inhibitors are being developed as cancer treatments. Some studies have demonstrated that MEK inhibitors show good treatment effects in ovarian cancer patients (Ref. Reference Shrestha158). Clinical trials NCT03363867 and NCT03363867 for the treatment of patients with ovarian cancer are ongoing.

Conclusion

The development of ICIs has revolutionised the management of many types of solid tumours, particularly advanced-stage cancers. Herein, we describe the mechanisms and expression of immune checkpoints and summarise recent updates regarding ICIs monotherapy or combined with other small-molecule-targeted agents in ovarian cancer. It is worth noting that the ICIs monotherapy have limited efficacy in ovarian cancer. Although synergistic therapies exhibit superior efficacy in patients with persistent or recurrent ovarian cancer compared with monotherapy, fundamental research and clinical use of combination therapy still encounters many obstacles, such as treatments with ICIs have limited response rates, no identified predictive biomarkers to select patients suited for ICIs, cannot effectively avoid the immune-related adverse events and the number of clinical trials of ICIs in ovarian cancer is relatively limited. Therefore, a more comprehensive understanding of immune checkpoints and immunosuppressive TME is crucial to improve the efficacy of ICIs in ovarian cancer.

Acknowledgements

We thank all authors who contributed to this article.

Conflict of interest

The authors have no other conflicts of interest to declare.

Ethical standards

The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

References

Siegel, RL et al. (2018) Cancer statistics. CA: A Cancer Journal for Clinicians 68, 730.Google ScholarPubMed
Torre, LA et al. (2018) Ovarian cancer statistics. CA: A Cancer Journal for Clinicians 68, 284296.Google ScholarPubMed
Hennessy, BT et al. (2009) Ovarian cancer. Lancet 374, 13711382.CrossRefGoogle ScholarPubMed
Siegel, RL et al. (2017) Cancer statistics. CA: A Cancer Journal for Clinicians 67, 730.Google ScholarPubMed
Odunsi, K (2017) Immunotherapy in ovarian cancer. Annals of Oncology 28, viii1viii7.CrossRefGoogle ScholarPubMed
Anadon, CM et al. (2022) Ovarian cancer immunogenicity is governed by a narrow subset of progenitor tissue-resident memory T cells. Cancer Cell 40, 545557.e513.CrossRefGoogle ScholarPubMed
Maiorano, BA et al. (2021) Ovarian cancer in the era of immune checkpoint inhibitors: state of the art and future perspectives. Cancers (Basel) 13, 4438.CrossRefGoogle ScholarPubMed
Yin, M et al. (2016) Tumor-associated macrophages drive spheroid formation during early transcoelomic metastasis of ovarian cancer. Journal of Clinical Investigation 126, 41574173.CrossRefGoogle ScholarPubMed
Kolomeyevskaya, N et al. (2015) Cytokine profiling of ascites at primary surgery identifies an interaction of tumor necrosis factor-α and interleukin-6 in predicting reduced progression-free survival in epithelial ovarian cancer. Gynecologic Oncology 138, 352357.CrossRefGoogle ScholarPubMed
Yuan, X et al. (2017) Prognostic significance of tumor-associated macrophages in ovarian cancer: a meta-analysis. Gynecologic Oncology 147, 181187.CrossRefGoogle ScholarPubMed
Curiel, TJ et al. (2004) Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nature Medicine 10, 942949.CrossRefGoogle ScholarPubMed
Edner, NM et al. (2020) Targeting co-stimulatory molecules in autoimmune disease. Nature Reviews. Drug Discovery 19, 860883.CrossRefGoogle ScholarPubMed
Jiang, X et al. (2019) Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Molecular Cancer 18, 10.CrossRefGoogle ScholarPubMed
Hirano, F et al. (2005) Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Research 65, 10891096.CrossRefGoogle ScholarPubMed
Okazaki, T et al. (2001) PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting Src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proceedings of the National Academy of Sciences of the USA 98, 1386613871.CrossRefGoogle ScholarPubMed
Egen, JG et al. (2002) CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nature Immunology 3, 611618.CrossRefGoogle ScholarPubMed
Lingel, H et al. (2019) CTLA-4 (CD152): a versatile receptor for immune-based therapy. Seminars in Immunology 42, 101298.CrossRefGoogle ScholarPubMed
Goldberg, MV et al. (2011) LAG-3 in cancer immunotherapy. Current Topics in Microbiology and Immunology 344, 269278.Google ScholarPubMed
Workman, CJ et al. (2004) Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. Journal of Immunology 172, 54505455.CrossRefGoogle ScholarPubMed
Workman, CJ et al. (2002) Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. Journal of Immunology 169, 53925395.CrossRefGoogle ScholarPubMed
Chiba, S et al. (2012) Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nature Immunology 13, 832842.CrossRefGoogle ScholarPubMed
Kang, CW et al. (2015) Apoptosis of tumor infiltrating effector TIM-3+CD8+ T cells in colon cancer. Scientific Reports 5, 15659.CrossRefGoogle ScholarPubMed
Murga-Zamalloa, CA et al. (2020) Expression of the checkpoint receptors LAG-3, TIM-3 and VISTA in peripheral T cell lymphomas. Journal of Clinical Pathology 73, 197203.CrossRefGoogle ScholarPubMed
Zhao, L et al. (2021) TIM-3: an update on immunotherapy. International Immunopharmacology 99, 107933.CrossRefGoogle ScholarPubMed
Chauvin, JM et al. (2020) TIGIT in cancer immunotherapy. Journal for Immunotherapy of Cancer 8, e000957.CrossRefGoogle ScholarPubMed
Dougall, WC et al. (2017) TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunological Reviews 276, 112120.CrossRefGoogle ScholarPubMed
Han, P et al. (2004) An inhibitory Ig superfamily protein expressed by lymphocytes and APCs is also an early marker of thymocyte positive selection. Journal of Immunology 172, 59315939.CrossRefGoogle ScholarPubMed
Sedy, JR et al. (2005) B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nature Immunology 6, 9098.CrossRefGoogle Scholar
Hosseinkhani, N et al. (2021) The role of V-domain Ig suppressor of T cell activation (VISTA) in cancer therapy: lessons learned and the road ahead. Frontiers in Immunology 12, 676181.CrossRefGoogle ScholarPubMed
Johnston, RJ et al. (2019) VISTA is an acidic pH-selective ligand for PSGL-1. Nature 574, 565570.CrossRefGoogle ScholarPubMed
Tang, K et al. (2021) Indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors in clinical trials for cancer immunotherapy. Journal of Hematology & Oncology 14, 68.CrossRefGoogle ScholarPubMed
Ishida, Y et al. (1992) Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO Journal 11, 38873895.CrossRefGoogle ScholarPubMed
Nishimura, H et al. (1999) Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 11, 141151.CrossRefGoogle ScholarPubMed
Marasco, M et al. (2020) Molecular mechanism of SHP2 activation by PD-1 stimulation. Science Advances 6, eaay4458.CrossRefGoogle ScholarPubMed
Terme, M et al. (2011) IL-18 induces PD-1-dependent immunosuppression in cancer. Cancer Research 71, 53935399.CrossRefGoogle ScholarPubMed
Matsuzaki, J et al. (2010) Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proceedings of the National Academy of Sciences of the USA 107, 78757880.CrossRefGoogle ScholarPubMed
Tu, L et al. (2020) Assessment of the expression of the immune checkpoint molecules PD-1, CTLA4, TIM-3 and LAG-3 across different cancers in relation to treatment response, tumor-infiltrating immune cells and survival. International Journal of Cancer 147, 423439.CrossRefGoogle ScholarPubMed
Rådestad, E et al. (2019) Immune profiling and identification of prognostic immune-related risk factors in human ovarian cancer. Oncoimmunology 8, e1535730.CrossRefGoogle ScholarPubMed
Hensler, M et al. (2020) M2-like macrophages dictate clinically relevant immunosuppression in metastatic ovarian cancer. Journal for Immunotherapy of Cancer 8, e000979.CrossRefGoogle ScholarPubMed
Maine, CJ et al. (2014) Programmed death ligand-1 over-expression correlates with malignancy and contributes to immune regulation in ovarian cancer. Cancer Immunology Immunotherapy 63, 215224.CrossRefGoogle ScholarPubMed
Abiko, K et al. (2013) PD-L1 on tumor cells is induced in ascites and promotes peritoneal dissemination of ovarian cancer through CTL dysfunction. Clinical Cancer Research 19, 13631374.CrossRefGoogle ScholarPubMed
Parvathareddy, SK et al. (2021) Differential expression of PD-L1 between primary and metastatic epithelial ovarian cancer and its clinico-pathological correlation. Scientific Reports 11, 3750.CrossRefGoogle ScholarPubMed
Darb-Esfahani, S et al. (2016) Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor-infiltrating lymphocytes in ovarian high grade serous carcinoma. Oncotarget 7, 14861499.CrossRefGoogle ScholarPubMed
Linsley, PS et al. (1991) CTLA-4 is a second receptor for the B cell activation antigen B7. Journal of Experimental Medicine 174, 561569.CrossRefGoogle ScholarPubMed
Krummel, MF et al. (1995) CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. Journal of Experimental Medicine 182, 459465.CrossRefGoogle ScholarPubMed
Duraiswamy, J et al. (2013) Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors. Cancer Research 73, 35913603.CrossRefGoogle ScholarPubMed
Huard, B et al. (1995) CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. European Journal of Immunology 25, 27182721.CrossRefGoogle ScholarPubMed
Grosso, JF et al. (2007) LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. Journal of Clinical Investigation 117, 33833392.CrossRefGoogle ScholarPubMed
Kouo, T et al. (2015) Galectin-3 shapes antitumor immune responses by suppressing CD8+ T cells via LAG-3 and inhibiting expansion of plasmacytoid dendritic cells. Cancer Immunology Research 3, 412423.CrossRefGoogle ScholarPubMed
Xu, F et al. (2014) LSECTin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Research 74, 34183428.CrossRefGoogle ScholarPubMed
Wang, J et al. (2019) Fibrinogen-like protein 1 is a major immune inhibitory ligand of LAG-3. Cell 176, 334347.e312.CrossRefGoogle Scholar
Woo, SR et al. (2012) Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Research 72, 917927.CrossRefGoogle ScholarPubMed
Huang, RY et al. (2015) LAG3 and PD1 co-inhibitory molecules collaborate to limit CD8+ T cell signaling and dampen antitumor immunity in a murine ovarian cancer model. Oncotarget 6, 2735927377.CrossRefGoogle Scholar
Das, M et al. (2017) TIM-3 and its role in regulating anti-tumor immunity. Immunological Reviews 276, 97111.CrossRefGoogle ScholarPubMed
Zhu, C et al. (2005) The TIM-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nature Immunology 6, 12451252.CrossRefGoogle ScholarPubMed
Santiago, C et al. (2007) Structures of T cell immunoglobulin mucin protein 4 show a metal-ion-dependent ligand binding site where phosphatidylserine binds. Immunity 27, 941951.CrossRefGoogle ScholarPubMed
Xu, Y et al. (2017) Role of TIM-3 in ovarian cancer. Clinical & Translational Oncology 19, 10791083.CrossRefGoogle ScholarPubMed
Yan, J et al. (2013) TIM-3 expression defines regulatory T cells in human tumors. PLoS ONE 8, e58006.CrossRefGoogle ScholarPubMed
Sakuishi, K et al. (2013) TIM3(+)FOXP3(+) regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer. Oncoimmunology 2, e23849.CrossRefGoogle ScholarPubMed
Whelan, S et al. (2019) PVRIG and PVRL2 are induced in cancer and inhibit CD8(+) T-cell function. Cancer Immunology Research 7, 257268.CrossRefGoogle ScholarPubMed
Li, YC et al. (2020) Overexpression of an immune checkpoint (CD155) in breast cancer associated with prognostic significance and exhausted tumor-infiltrating lymphocytes: a cohort study. Journal of Immunology Research 2020, 3948928.CrossRefGoogle ScholarPubMed
Harjunpää, H et al. (2020) TIGIT as an emerging immune checkpoint. Clinical and Experimental Immunology 200, 108119.CrossRefGoogle ScholarPubMed
Maas, RJ et al. (2020) TIGIT blockade enhances functionality of peritoneal NK cells with altered expression of DNAM-1/TIGIT/CD96 checkpoint molecules in ovarian cancer. Oncoimmunology 9, 1843247.CrossRefGoogle ScholarPubMed
Chen, F et al. (2020) TIGIT enhances CD4(+) regulatory T-cell response and mediates immune suppression in a murine ovarian cancer model. Cancer Medicine 9, 35843591.CrossRefGoogle Scholar
Yu, X et al. (2019) BTLA/HVEM signaling: milestones in research and role in chronic hepatitis B virus infection. Frontiers in Immunology 10, 617.CrossRefGoogle ScholarPubMed
Zhang, T et al. (2016) Knockdown of HVEM, a lymphocyte regulator gene, in ovarian cancer cells increases sensitivity to activated T cells. Oncology Research 24, 189196.CrossRefGoogle ScholarPubMed
Chen, YL et al. (2019) BTLA blockade enhances cancer therapy by inhibiting IL-6/IL-10-induced CD19(high) B lymphocytes. Journal for Immunotherapy of Cancer 7, 313.CrossRefGoogle ScholarPubMed
Lines, JL et al. (2014) VISTA is an immune checkpoint molecule for human T cells. Cancer Research 74, 19241932.CrossRefGoogle ScholarPubMed
ElTanbouly, MA et al. (2020) VISTA: coming of age as a multi-lineage immune checkpoint. Clinical and Experimental Immunology 200, 120130.CrossRefGoogle ScholarPubMed
Mulati, K et al. (2019) VISTA expressed in tumour cells regulates T cell function. British Journal of Cancer 120, 115127.CrossRefGoogle ScholarPubMed
Zong, L et al. (2020) VISTA expression is associated with a favorable prognosis in patients with high-grade serous ovarian cancer. Cancer Immunology Immunotherapy 69, 3342.CrossRefGoogle ScholarPubMed
Liu, M et al. (2018) Targeting the IDO1 pathway in cancer: from bench to bedside. Journal of Hematology & Oncology 11, 100.CrossRefGoogle ScholarPubMed
Wainwright, DA et al. (2012) IDO expression in brain tumors increases the recruitment of regulatory T cells and negatively impacts survival. Clinical Cancer Research 18, 61106121.CrossRefGoogle ScholarPubMed
Mellor, AL et al. (2002) Cells expressing indoleamine 2,3-dioxygenase inhibit T cell responses. Journal of Immunology 168, 37713776.CrossRefGoogle ScholarPubMed
Qian, F et al. (2009) 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. Cancer Research 69, 54985504.CrossRefGoogle ScholarPubMed
Okamoto, A et al. (2005) Indoleamine 2,3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells. Clinical Cancer Research 11, 60306039.CrossRefGoogle ScholarPubMed
Takao, M et al. (2007) Increased synthesis of indoleamine-2,3-dioxygenase protein is positively associated with impaired survival in patients with serous-type, but not with other types of, ovarian cancer. Oncology Reports 17, 13331339.Google Scholar
Amobi-McCloud, A et al. (2021) IDO1 expression in ovarian cancer induces PD-1 in T cells via aryl hydrocarbon receptor activation. Frontiers in Immunology 12, 678999.CrossRefGoogle Scholar
Boland, JL et al. (2019) Early disease progression and treatment discontinuation in patients with advanced ovarian cancer receiving immune checkpoint blockade. Gynecologic Oncology 152, 251258.CrossRefGoogle ScholarPubMed
Wolchok, JD et al. (2010) Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study. The Lancet. Oncology 11, 155164.CrossRefGoogle ScholarPubMed
Hodi, FS et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. New England Journal of Medicine 363, 711723.CrossRefGoogle ScholarPubMed
Zamarin, D et al. (2020) Randomized phase II trial of nivolumab versus nivolumab and ipilimumab for recurrent or persistent ovarian cancer: an NRG oncology study. Journal of Clinical Oncology 38, 18141823.CrossRefGoogle ScholarPubMed
Hamanishi, J et al. (2015) Safety and antitumor activity of anti-PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer. Journal of Clinical Oncology 33, 40154022.CrossRefGoogle ScholarPubMed
Hamanishi, J et al. (2021) Nivolumab versus gemcitabine or pegylated liposomal doxorubicin for patients with platinum-resistant ovarian cancer: open-label, randomized trial in Japan (NINJA). Journal of Clinical Oncology 39, 36713681.CrossRefGoogle ScholarPubMed
Matulonis, UA et al. (2019) Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study. Annals of Oncology 30, 10801087.CrossRefGoogle ScholarPubMed
Walsh, CS et al. (2021) Phase II trial of cisplatin, gemcitabine and pembrolizumab for platinum-resistant ovarian cancer. PLoS ONE 16, e0252665.CrossRefGoogle ScholarPubMed
Lee, EK et al. (2020) Combined pembrolizumab and pegylated liposomal doxorubicin in platinum resistant ovarian cancer: a phase 2 clinical trial. Gynecologic Oncology 159, 7278.CrossRefGoogle ScholarPubMed
Varga, A et al. (2019) Pembrolizumab in patients with programmed death ligand 1-positive advanced ovarian cancer: analysis of KEYNOTE-028. Gynecologic Oncology 152, 243250.CrossRefGoogle ScholarPubMed
Nishio, S et al. (2020) Pembrolizumab monotherapy in Japanese patients with advanced ovarian cancer: subgroup analysis from the KEYNOTE-100. Cancer Science 111, 13241332.CrossRefGoogle ScholarPubMed
Rahma, OE et al. (2022) Phase IB study of ziv-aflibercept plus pembrolizumab in patients with advanced solid tumors. Journal for Immunotherapy of Cancer 10, e003569.CrossRefGoogle ScholarPubMed
Brahmer, JR et al. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. New England Journal of Medicine 366, 24552465.CrossRefGoogle ScholarPubMed
Taylor, K et al. (2020) An open-label, phase II multicohort study of an oral hypomethylating agent CC-486 and durvalumab in advanced solid tumors. Journal for Immunotherapy of Cancer 8, e000883.CrossRefGoogle ScholarPubMed
Ngoi, NY et al. (2020) A multicenter phase II randomized trial of durvalumab (MEDI-4736) versus physician's choice chemotherapy in recurrent ovarian clear cell adenocarcinoma (MOCCA). International Journal of Gynecological Cancer 30, 12391242.CrossRefGoogle ScholarPubMed
Lee, JY et al. (2019) A phase II study of neoadjuvant chemotherapy plus durvalumab and tremelimumab in advanced-stage ovarian cancer: a Korean gynecologic oncology group study (KGOG 3046), TRU-D. Journal of Gynecologic Oncology 30, e112.CrossRefGoogle Scholar
Liu, JF et al. (2019) Safety, clinical activity and biomarker assessments of atezolizumab from a phase I study in advanced/recurrent ovarian and uterine cancers. Gynecologic Oncology 154, 314322.CrossRefGoogle ScholarPubMed
Pujade-Lauraine, E et al. (2021) Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum-resistant or platinum-refractory ovarian cancer (JAVELIN ovarian 200): an open-label, three-arm, randomised, phase 3 study. The Lancet. Oncology 22, 10341046.CrossRefGoogle ScholarPubMed
Pujade-Lauraine, E et al. (2018) Avelumab (anti-PD-L1) in platinum-resistant/refractory ovarian cancer: JAVELIN ovarian 200 phase III study design. Future Oncology 14, 21032113.CrossRefGoogle ScholarPubMed
Monk, BJ et al. (2021) Chemotherapy with or without avelumab followed by avelumab maintenance versus chemotherapy alone in patients with previously untreated epithelial ovarian cancer (JAVELIN ovarian 100): an open-label, randomised, phase 3 trial. The Lancet. Oncology 22, 12751289.CrossRefGoogle ScholarPubMed
Disis, ML et al. (2019) Efficacy and safety of avelumab for patients with recurrent or refractory ovarian cancer: phase 1b results from the JAVELIN solid tumor trial. JAMA Oncology 5, 393401.CrossRefGoogle ScholarPubMed
Ansell, SM et al. (2009) Phase I study of ipilimumab, an anti-CTLA-4 monoclonal antibody, in patients with relapsed and refractory B-cell non-Hodgkin lymphoma. Clinical Cancer Research 15, 64466453.CrossRefGoogle ScholarPubMed
Normann, MC et al. (2019) Early experiences with PD-1 inhibitor treatment of platinum resistant epithelial ovarian cancer. Journal of Gynecologic Oncology 30, e56.CrossRefGoogle ScholarPubMed
Linette, GP et al. (2019) Tumor-infiltrating lymphocytes in the checkpoint inhibitor era. Current Hematologic Malignancy Reports 14, 286291.CrossRefGoogle ScholarPubMed
Cheng, WC et al. (2019) Firing up cold tumors. Trends in Cancer 5, 528530.CrossRefGoogle ScholarPubMed
Bezu, L et al. (2015) Combinatorial strategies for the induction of immunogenic cell death. Frontiers in Immunology 6, 187.Google ScholarPubMed
Lheureux, S et al. (2020) EVOLVE: a multicenter open-label single-arm clinical and translational phase II trial of cediranib plus olaparib for ovarian cancer after PARP inhibition progression. Clinical Cancer Research 26, 42064215.CrossRefGoogle ScholarPubMed
Liu, JF et al. (2019) Assessment of combined nivolumab and bevacizumab in relapsed ovarian cancer: a phase 2 clinical trial. JAMA Oncology 5, 17311738.CrossRefGoogle ScholarPubMed
Zsiros, E et al. (2021) Efficacy and safety of pembrolizumab in combination with bevacizumab and oral metronomic cyclophosphamide in the treatment of recurrent ovarian cancer: a phase 2 nonrandomized clinical trial. JAMA Oncology 7, 7885.CrossRefGoogle ScholarPubMed
Moore, KN et al. (2021) Atezolizumab, bevacizumab, and chemotherapy for newly diagnosed stage III or IV ovarian cancer: placebo-controlled randomized phase III trial (IMagyn050/GOG 3015/ENGOT-OV39). Journal of Clinical Oncology 39, 18421855.CrossRefGoogle ScholarPubMed
Moroney, JW et al. (2020) Safety and clinical activity of atezolizumab plus bevacizumab in patients with ovarian cancer: a phase Ib study. Clinical Cancer Research 26, 56315637.CrossRefGoogle ScholarPubMed
Gonzalez Martin, A et al. (2021) A phase III, randomized, double blinded trial of platinum based chemotherapy with or without atezolizumab followed by niraparib maintenance with or without atezolizumab in patients with recurrent ovarian, tubal, or peritoneal cancer and platinum treatment free interval of more than 6 months: ENGOT-Ov41/GEICO 69-O/ANITA trial. International Journal of Gynecological Cancer 31, 617622.CrossRefGoogle ScholarPubMed
Lee, YJ et al. (2021) A single-arm phase II study of olaparib maintenance with pembrolizumab and bevacizumab in BRCA non-mutated patients with platinum-sensitive recurrent ovarian cancer (OPEB-01). Journal of Gynecologic Oncology 32, e31.CrossRefGoogle ScholarPubMed
Lampert, EJ et al. (2020) Combination of PARP inhibitor olaparib, and PD-L1 inhibitor durvalumab, in recurrent ovarian cancer: a proof-of-concept phase II study. Clinical Cancer Research 26, 42684279.CrossRefGoogle ScholarPubMed
Fumet, JD et al. (2020) Precision medicine phase II study evaluating the efficacy of a double immunotherapy by durvalumab and tremelimumab combined with olaparib in patients with solid cancers and carriers of homologous recombination repair genes mutation in response or stable after olaparib treatment. BMC Cancer 20, 748.CrossRefGoogle ScholarPubMed
Zimmer, AS et al. (2019) A phase I study of the PD-L1 inhibitor, durvalumab, in combination with a PARP inhibitor, olaparib, and a VEGFR1-3 inhibitor, cediranib, in recurrent women's cancers with biomarker analyses. Journal for Immunotherapy of Cancer 7, 197.CrossRefGoogle Scholar
Lee, JY et al. (2022) Biomarker-guided targeted therapy in platinum-resistant ovarian cancer (AMBITION; KGOG 3045): a multicentre, open-label, five-arm, uncontrolled, umbrella trial. Journal of Gynecologic Oncology 33, e45.CrossRefGoogle ScholarPubMed
Monk, BJ et al. (2021) ATHENA (GOG-3020/ENGOT-ov45): a randomized, phase III trial to evaluate rucaparib as monotherapy (ATHENA-MONO) and rucaparib in combination with nivolumab (ATHENA-COMBO) as maintenance treatment following frontline platinum-based chemotherapy in ovarian cancer. International Journal of Gynecological Cancer 31, 15891594.CrossRefGoogle ScholarPubMed
Jung, KH et al. (2019) Phase I study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) administered with PD-L1 inhibitor (atezolizumab) in advanced solid tumors. Clinical Cancer Research 25, 32203228.CrossRefGoogle Scholar
Chen, S et al. (2022) Epigenetic priming enhances antitumor immunity in platinum-resistant ovarian cancer. Journal of Clinical Investigation 132, e158800.CrossRefGoogle ScholarPubMed
Zamarin, D et al. (2020) Safety, immunogenicity, and clinical efficacy of durvalumab in combination with folate receptor alpha vaccine TPIV200 in patients with advanced ovarian cancer: a phase II trial. Journal for Immunotherapy of Cancer 8, e000829.CrossRefGoogle ScholarPubMed
Falchook, GS et al. (2021) A phase 1a/1b trial of CSF-1R inhibitor LY3022855 in combination with durvalumab or tremelimumab in patients with advanced solid tumors. Investigational New Drugs 39, 12841297.CrossRefGoogle ScholarPubMed
Rocconi, RP et al. (2022) Proof of principle study of sequential combination atezolizumab and Vigil in relapsed ovarian cancer. Cancer Gene Therapy 29, 369382.CrossRefGoogle ScholarPubMed
Simonelli, M et al. (2022) Isatuximab plus atezolizumab in patients with advanced solid tumors: results from a phase I/II, open-label, multicenter study. ESMO Open 7, 100562.CrossRefGoogle ScholarPubMed
Li, J et al. (2019) Expanding the role of STING in cellular homeostasis and transformation. Trends in Cancer 5, 195197.CrossRefGoogle ScholarPubMed
Curdy, N et al. (2019) Regulatory mechanisms of inhibitory immune checkpoint receptors expression. Trends in Cell Biology 29, 777790.CrossRefGoogle ScholarPubMed
Huang, RY et al. (2017) Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology 6, e1249561.CrossRefGoogle ScholarPubMed
Venkitaraman, AR (2002) Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell 108, 171182.CrossRefGoogle ScholarPubMed
Konstantinopoulos, PA et al. (2015) Homologous recombination deficiency: exploiting the fundamental vulnerability of ovarian cancer. Cancer Discovery 5, 11371154.CrossRefGoogle ScholarPubMed
Pilié, PG et al. (2019) State-of-the-art strategies for targeting the DNA damage response in cancer. Nature Reviews. Clinical Oncology 16, 81104.CrossRefGoogle ScholarPubMed
Foo, T et al. (2021) PARP inhibitors in ovarian cancer: an overview of the practice-changing trials. Genes Chromosomes & Cancer 60, 385397.CrossRefGoogle ScholarPubMed
Zheng, F et al. (2020) Mechanism and current progress of poly ADP-ribose polymerase (PARP) inhibitors in the treatment of ovarian cancer. Biomedicine & Pharmacotherapy 123, 109661.CrossRefGoogle ScholarPubMed
da Cunha Colombo Bonadio, RR et al. (2018) Homologous recombination deficiency in ovarian cancer: a review of its epidemiology and management. Clinics (Sao Paulo) 73, e450s.CrossRefGoogle ScholarPubMed
Ding, L et al. (2018) PARP inhibition elicits STING-dependent antitumor immunity in BRCA1-deficient ovarian cancer. Cell Reports 25, 29722980.e2975.CrossRefGoogle ScholarPubMed
Wang, Z et al. (2019) Niraparib activates interferon signaling and potentiates anti-PD-1 antibody efficacy in tumor models. Scientific Reports 9, 1853.CrossRefGoogle ScholarPubMed
Shen, J et al. (2019) PARPi triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCAness. Cancer Research 79, 311319.CrossRefGoogle ScholarPubMed
Konstantinopoulos, PA et al. (2019) Single-arm phases 1 and 2 trial of niraparib in combination with pembrolizumab in patients with recurrent platinum-resistant ovarian carcinoma. JAMA Oncology 5, 11411149.CrossRefGoogle ScholarPubMed
Bekes, I et al. (2016) Does VEGF facilitate local tumor growth and spread into the abdominal cavity by suppressing endothelial cell adhesion, thus increasing vascular peritoneal permeability followed by ascites production in ovarian cancer? Molecular Cancer 15, 13.CrossRefGoogle ScholarPubMed
Nagy, JA et al. (1995) Pathogenesis of ascites tumor growth: vascular permeability factor, vascular hyperpermeability, and ascites fluid accumulation. Cancer Research 55, 360368.Google ScholarPubMed
Luo, JC et al. (1998) Significant expression of vascular endothelial growth factor/vascular permeability factor in mouse ascites tumors. Cancer Research 58, 26522660.Google ScholarPubMed
Mahner, S et al. (2010) TIMP-1 and VEGF-165 serum concentration during first-line therapy of ovarian cancer patients. BMC Cancer 10, 139.CrossRefGoogle ScholarPubMed
Azam, F et al. (2010) Mechanisms of resistance to antiangiogenesis therapy. European Journal of Cancer 46, 13231332.CrossRefGoogle ScholarPubMed
Bergers, G et al. (2008) Modes of resistance to anti-angiogenic therapy. Nature Reviews Cancer 8, 592603.CrossRefGoogle ScholarPubMed
Yang, Y et al. (2021) Programmed death ligand-1 regulates angiogenesis and metastasis by participating in the c-JUN/VEGFR2 signaling axis in ovarian cancer. Cancer Communications 41, 511527.CrossRefGoogle ScholarPubMed
Klose, RJ et al. (2006) Genomic DNA methylation: the mark and its mediators. Trends in Biochemical Sciences 31, 8997.CrossRefGoogle ScholarPubMed
Ahluwalia, A et al. (2001) DNA methylation in ovarian cancer. II. Expression of DNA methyltransferases in ovarian cancer cell lines and normal ovarian epithelial cells. Gynecologic Oncology 82, 299304.CrossRefGoogle ScholarPubMed
Stone, TW et al. (2013) An expanding range of targets for kynurenine metabolites of tryptophan. Trends in Pharmacological Sciences 34, 136143.CrossRefGoogle ScholarPubMed
Li, F et al. (2017) IDO1: an important immunotherapy target in cancer treatment. International Immunopharmacology 47, 7077.CrossRefGoogle ScholarPubMed
Choi, SW et al. (2000) Folate and carcinogenesis: an integrated scheme. Journal of Nutrition 130, 129132.CrossRefGoogle ScholarPubMed
Scaranti, M et al. (2020) Exploiting the folate receptor α in oncology. Nature Reviews. Clinical Oncology 17, 349359.CrossRefGoogle ScholarPubMed
Figini, M et al. (2003) Reversion of transformed phenotype in ovarian cancer cells by intracellular expression of anti folate receptor antibodies. Gene Therapy 10, 10181025.CrossRefGoogle ScholarPubMed
Nawaz, FZ et al. (2022) Emerging roles for folate receptor FOLR1 in signaling and cancer. Trends in Endocrinology and Metabolism 33, 159174.CrossRefGoogle ScholarPubMed
Wade, PA et al. (1997) Histone acetylation: chromatin in action. Trends in Biochemical Sciences 22, 128132.CrossRefGoogle ScholarPubMed
Eckschlager, T et al. (2017) Histone deacetylase inhibitors as anticancer drugs. International Journal of Molecular Sciences 18, 1414.CrossRefGoogle ScholarPubMed
Li, Y et al. (2016) HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harbor Perspectives in Medicine 6, a026831.CrossRefGoogle ScholarPubMed
Weichert, W et al. (2008) Expression of class I histone deacetylases indicates poor prognosis in endometrioid subtypes of ovarian and endometrial carcinomas. Neoplasia (New York, N.Y.) 10, 10211027.CrossRefGoogle ScholarPubMed
Yano, M et al. (2018) Association of histone deacetylase expression with histology and prognosis of ovarian cancer. Oncology Letters 15, 35243531.Google ScholarPubMed
Roberts, PJ et al. (2007) Targeting the Raf–MEK–ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 26, 32913310.CrossRefGoogle ScholarPubMed
De Luca, A et al. (2012) The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: role in cancer pathogenesis and implications for therapeutic approaches. Expert Opinion on Therapeutic Targets 16(suppl. 2), S17S27.CrossRefGoogle ScholarPubMed
Shrestha, R et al. (2021) Multiomics characterization of low-grade serous ovarian carcinoma identifies potential biomarkers of MEK inhibitor sensitivity and therapeutic vulnerability. Cancer Research 81, 16811694.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Neo-antigens derived from tumour cells to CTL through MHC class I–TCRs and a co-stimulation signal of CD80 and/or CD86–CD28 interactions, CTLs are subsequently activated to destroy tumour cells. However, tumour cells often escape immune destruction through upregulation of immune checkpoint ligands, such as programmed cell death 1 ligand 1 (PD- L1), that can bind the immune checkpoint receptors programmed cell death 1 (PD-1) on the CTLs to deliver suppressing signals, finally inhibit the proliferation and activation of CTLs. Another negative-regulate immune checkpoint molecule cytotoxic T lymphocyte protein 4 (CTLA-4) that binds CD80 and CD86 and prevents their interaction with CD28, inhibit the co-stimulation signal of CD80 and/or CD86-CD28 interactions, thus inhibit the proliferation and activation of CTLs. ICIs could effectively prevent this effect. ICIs highly specifically bind to immune checkpoints, blocking this inhibitory mechanism and thereby reactivating the anti-tumour immune response.

Figure 1

Table 1. Summary of immune checkpoints and functions

Figure 2

Table 2. Clinical trials of ICI monotherapy in ovarian cancer

Figure 3

Table 3. Clinical trials of ICIs combined with other small-molecule-targeted agents in ovarian cancer