Skip to main content

Immunotherapy in endometrial cancer: rationale, practice and perspectives

Abstract

Tumor immunotherapy has attracted more and more attention nowadays, and multiple clinical trials have confirmed its effect in a variety of solid tumors. Immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell transfer (ACT), and lymphocyte-promoting cytokines are the main immunotherapy methods. Endometrial cancer (EC) is one of the most frequent tumors in women and the prognosis of recurrent or metastatic EC is poor. Since molecular classification has been applied to EC, immunotherapy for different EC subtypes (especially POLE and MSI-H) has gradually attracted attention. In this review, we focus on the expression and molecular basis of the main biomarkers in the immunotherapy of EC firstly, as well as their clinical application significance and limitations. Blocking tumor immune checkpoints is one of the most effective strategies for cancer treatment in recent years, and has now become the focus in the field of tumor research and treatment. We summarized clinical date of planned and ongoing clinical trials and introduced other common immunotherapy methods in EC, such as cancer vaccine and ACT. Hormone aberrations, metabolic syndrome (MetS) and p53 mutant and that affect the immunotherapy of endometrial cancer will also be discussed in this review.

Background

Uterine cancer, primarily EC, is the most common gynecologic tumor in developed countries [1] and ranks as the fourth highest among all cancers in number of estimated new cases among American females in 2020 [2]. The incidence rate of EC increased continuously (1.3% per year from 2007-2016) [3] partly due to the recent rise of nonendometrioid cases [4]. Most patients with EC are diagnosed at stage I because of notable early symptoms (irregular vaginal bleeding) [1]. Surgery including total hysterectomy and bilateral salpingo-oopherectomy with surgical staging is recommended as the standard management for medically operable patients, and adjuvant therapy (chemotherapy or radiotherapy) determinations are tailored on the basis of risk factors and pathologic findings, including age, tumor size and grade, lymphovascular invasion,lymph node involvement, degree of myometrium invasion, and lower uterine segment involvement. Among EC patients, 66.9% are diagnosed at local stage with a 5-year survival rate of 95.0%, conferring a good prognosis. However, 16% of EC patients have a metastatic disease with 5-year survival rate of 16.8%, which contributes disproportionally to disease mortality [5]. For those patients with uterine confined lesions and high-intermediate risk for recurrence, adjuvant radiotherapy (RT) is recommended. Carboplatin/paclitaxel systemic chemotherapy and RT can benefit those with high risk for recurrence. As for the patients with recurrent, extra-uterine lesions, systemic, combination therapy with or without RT is considered as suitable. However, effective treatment options are lacking for patients with advanced disease following standard therapy.

Immunotherapy has been recognized as a new, powerful approach for a variety of human carcinomas, with considerable clinical response seen in a portion of recurrent or refractory cases [6,7,8]. There are few second-line treatment options for endometrial cancer, and some progress has been made in immunotherapy in recent years. In March 2020, the national comprehensive cancer network (NCCN) recommended The Cancer Genome Atlas Research Network (TCGA) molecular typing of endometrial cancer for the first time and included it in the guidelines for diagnosis and treatment of endometrial cancer, which indicates the era of immunotherapy based on tumor microenvironment and genotype. With the continuous elucidation of the pathogenesis of endometrial cancer, more and more evidence shows that a large number of immune cells and cytokines can be seen in endometrial cancer tissue, and can stimulate endogenous anti-tumor immune response. Compared with other gynecological malignant tumors, endometrial cancer is most likely to benefit from immunotherapy [9,10,11]. The rationale for cancer immunotherapy can be summarized as reversing the tumors immuno-suppressive effects of the tumors or enhancing the inherent anti-tumor immune responses of the host [12]. The main cancer immunotherapy approaches include ICIs, cancer vaccines, ACT, and lymphocyte-promoting cytokines. Several methods such as oncolytic viruses and bispecific antibodies [13] are also emerging. Immunotherapy in EC, especially in those with advanced or metastatic disease, is drawing intense attention currently, as more than 50 clinical trials investigating various categories of immunotherapy in EC have been listed on the clinicaltrials.gov website (Table 1). For targeted therapy, the significance of biomarker's (Table 2) detection and clinical guidance is more clear. With the gradual application of immunotherapy in the clinic, more effort will be needed in this regard.

Table 1 Published or ongoing trials of immunotherapy in EC:
Table 2 Biomarkers in ECs

In 2013, TCGA [14] published an article regarding the integrated genomic classification of EC, classifying EC into four distinct molecular subtypes: Polymerase-ε(POLE) ultramutated, microsatellite instability hypermutated (MSI-H), copy-number low and copy-number high. The characteristics of these four subtypes which were determined through somatic mutations, microsatellite status and copy number alterations [14] are listed as follows:

POLE ultramutated ECs harbor DNA somatic mutations in the exonuclease domain within POLE gene and account for 7-12% of total EC patients [15, 16]. This subtype has a remarkably high mutation rate of 232 × 10–6 mutations/Mb. Patients of this subtype, though sometimes having high histological grades [17], usually have a good prognosis [16, 18,19,20], perhaps because of high immunogenicity [21] and chemosensitivity [22] due to DNA repair deficiency.

The MSI-hypermutated type of EC has diverse lengths of DNA microsatellites [14, 23, 24] in the genome and is also characterized by a high mutation rate (18×10-6mutations/Mb). Deficiency in the DNA mismatch repair (MMR) system, which is commonly due to gene mutations in MLH1, MSH2, MSH6 or PMS2, is responsible for the phenotype of MSI EC. Somatic mutations of these four MMR-related genes, usually, MLH1 promoter methylation [23], are often detected in MSI sporadic EC, whereas germ-line mutations of MMR-related genes are mainly found in hereditary EC, such as LS patients [25]. There is no statistically significant correlation between MSI status and clinical outcome [26, 27].

Both POLE and MSI subtypes of EC are generally considered as having high genome instability and immunogenic phenotypes [28, 29], with overexpressed immune-related biomarkers which will be further described in the following content.

Copy-number low ECs are also defined as microsatellite stability (MSS) ECs and have a low mutation rate (2.9 × 10–6 mutations/Mb). This type of EC accounts for nearly 60% of low-grade EC cases in TCGA, while only accounting for 8.7% of high grade ECs. Mutant PTEN and PIK3CA genes are found in 77% and 53% of this type of ECs respectively [14, 30]. Prognosis of the copy-number low subtype overlaps with MSI-H subtype with no obvious relationship between this subtype and outcomes [31].

The copy-number high subgroup is principally composed of serous and mixed histology tumors, with a residual portion of high grade endometrioid ECs. TP53 mutation is generally detected (92%) in this type of EC, while KRAS and PTEN mutations infrequently occur. The prognosis of patients with this type of EC is poor and is worsened by unfavorable clinicopathological characteristics [24, 30,31,32]. Copy-number high/serous-like EC shares similar molecular patterns with high-grade serous ovarian carcinoma (HGSOC) and basal-like breast carcinoma, including highly mutant TP53 gene (84%~96%), a low frequency of PTEN mutations (1%~2%) and similar focal somatic copy number alterations [14]. Both copy-number low and Copy-number high ECs are conventionally considered hypomutated ECs, characterized by low expression of immune-related biomarkers.

Immune system in normal endometrium

The immune system in normal endometrium is mainly composed of endometrial epithelial cells, innate and adaptive immune cells, and inflammatory cytokines [33]. The endometrial epithelial cells, also a component of the mucosal immune system, have the functions of providing physical barriers [33], secreting defensins [34] or other immune mediators [35], and presenting antigens [36]. Both innate immune cells and adaptive immune cells participate in common surveillance and pathogen elimination in normal endometrium [33]. The inflammatory cytokines can recruit immune migratory cells [37] and modulate the immune network of the endometrium [33]. All these components’ functions are precisely regulated by sex hormones, such as estradiol and progesterone, and vary depending on the changes of menstrual cycles [33, 37, 38]. Macrophages and neutrophils are at the highest levels prior to menstruation [33] at which time they partly participate in the immunological protection and destruction of endometrial tissue [39]. Natural killer (NK) cells are inert during normal menstrual cycles and can mature to functional ones during pregnancy [40]. Regulatory T cells (Treg) can suppress ongoing immune responses in the endometrial microenvironment. T and B cells exist as aggregates in uterine mucosa [41,42,43], which are hormone-driven and absent in menopause [41]. The number of NK cells, Tregs, and T/B cell aggregates are increased during the menstrual cycle [33, 44], suggesting complicated immune modulation. Endometrial immunity, including all these components, plays a dual role in normal physiological processes by creating an immunosuppressive environment to avoid feto-maternal rejection while also protecting the disrupted endometrium from pathogens during menstruation [33].

Rationale of Tumor-immunity interaction in EC

When the endometrium becomes cancerous, the immune microenvironment appears different from that of the normal endometrium. Endogenous or exogenous cancerogenic factors can directly modulate immune related signaling pathways or the host's defensive inflammation and also indirectly change the initial immune balance by tumor induced immunoediting. Endometrial immunity plays a paradoxical role during carcinogenesis, having both anti-tumor and tumor-promoting effects.

In the late 50’s, Burnet and Thomas introduced the “cancer immunosurveillance” hypothesis, which described that in immunocompetent hosts, tumor elicited a response from the immune system to evolve to control the malignant cell outgrowth [45, 46]. This conceptual model built a theoretical relationship between the immune system and tumor growth. However, controversy persisted with the results from several experimental studies [47, 48] providing little support for this hypothesis. The cancer immunosurveillance hypothesis was gradually abandoned. In 2002, Ikeda, Old and Schreiber [45, 49] first postulated a more complex “Cancer Immunoediting” model in which the immune system could both constrain and promote tumor development. It was defined by three distinct phases: elimination, equilibrium and escape. The tumor-immunity reciprocal activities in EC will be illustrated according to this classical model.

First, in the elimination phase, both innate and adaptive immune response are involved in the identification and cytotoxic elimination of EC cells [50]. Under stressful and dangerous conditions, the EC cells represent “altered self” phenotypes and express “non-self” antigens [51], which are phagocytized and processed by the dendritic cells (DCs) [52]. DCs are primed and then present these tumor-associated antigens to generate T cell responses including the production of CD8+ cytotoxic T cells (CTLs) and CD4+ T cells [52, 53]. CD8+ CTLs can directly kill EC cells while CD4+ T helper cells elicit a specific B cell response to provide both humoral and cytotoxic immune responses [54]. If the immune system completely wipes out the EC cells, the elimination phase can be termed as the endpoint of the cancer immunoediting process [55]. However, rare residual malignant cells may survive the elimination phase and enter the equilibrium phase. During the “Equilibrium” phase, the EC cells and immune system shape each other, and a temporal biological balance is established [56]. Several latent EC cells can reside in patients for decades and are maintained in a state of dormancy [57]. In this period, a complex interplay occurs between the elements of the immune system and EC, which will define the final outcome of the tumor’s existence. If the EC cells sculpt the immune system to produce an immunosuppressed environment, they will then escape the immunologic control and enter into the “Escape” phase [58]. The ECs resume growth and even forms distant metastases [55].

Understanding the mechanism underlying the transition of equilibrium phase to escape phase can help to develop immunotherapies. Researchers have now identified that the tumor cells secrete vascular endothelial growth factor, transforming growth factor-β, and indoleamine 2, 3-dioxygenase ( IDO) to inactivate or dampen immune cells [59,60,61]. Tumor cells can also escape immunosurveillance by losing the expression of tumor antigen and/or the major histocompatibility complex class I molecules [62] or through the immuno-inhibitory effects exerted by Treg and myeloid derived suppressor cells (MDSC). The up-regulation of immune checkpoints (Table 3), which block activated T cells through inhibitory pathways, is plays a crucial role in evading immune-surveillance [63]. In EC, when the immune system attempts to recognize and remove the tumor cells, two signals are necessary for enabling naïve T cell activation [64]. The first signal is the binding of T cell receptors (TCR) on T cells to antigenic peptide-bearing major histocompatibility complex on EC cells [55, 65], which alone is insufficient for T cell stimulation. The second signal is generated through the binding of costimulatory molecules, such as CD80 and CD86 (also known as B7-1 and B7-2), on the antigen-presenting cell (APC) to corresponding ligands (e.g. CD28) on the T cell [66]. This two-signal activation process can be negatively regulated by immune checkpoint pathways that can be exploited by EC cells to eliminate immune attack [67,68,69]. Programmed cell death 1 (PD1) and cytotoxic T lymphocyte antigen 4 (CTLA4) expressed on T or other immune cells and programmed cell death 1 ligand 1 (PDL1) expressed on EC cells are all principal components involved in immune checkpoint signaling. CTLA4 dampens costimulatory signaling of the CD28/B7 axis (the second signal) via competition with CD28 for binding to B7 ligands [69,70,71]. PD-1/PD-L1 engagement recruits tyrosine phosphatase SHP2 which dephosphorylates proximal signaling elements of T cell receptors signaling [72], thus producing a negative costimulatory effect and dampening T cell activation through T cell receptors signaling interference [69]. These immune checkpoint signals are defined as key targets for recently developed immunotherapies.

Table 3 Immune checkpoints in ECs

Biomarkers for EC immunotherapy

PD-L1

PD-L1 is one of several ligands for the PD-1 receptor. Tumor cells can up-regulate the expression of PD-L1 which binds to PD-1 on T cells activating the co-inhibitory signal in T cells and thus avoiding T cell cytolysis and facilitating tumor progression [73,74,75]. This pathway is one of the main targets of ICIs. Recent research has found that anti-PD-1/PD-L1 therapy has response rates of 36-100% in PD-L1-positive tumors compared with only response rates of 0–17% in PD-L1-negative tumors across all tumor types [75]. The expression of PD-L1 in the tumor microenvironment has been recognized as an important biomarker by demonstrating which patients are more likely to benefit from immunotherapy.

ECs show 75% expression of PD-1 and 25–100% expression of PD-L1, which are both the highest levels among gynecological cancers. However, PD-L1 expression patterns among different molecular subtypes of EC are controversial. In 2015, Howitt et al. [28] evaluated PD-L1 expression in 63 patients with EC. PD-L1 expression was more frequent in POLE and MSI tumors compared with MSS tumors in both intraepithelial immune cells based on presence vs total absence (P=0.02) and in peritumoral immune cells based on at least 10% expression (84% vs 56%, P=0.03), PD-L1 expression did not show significant difference in tumor cells among POLE, MSI, and MSS ECs. The higher expression levels of PD-L1 in POLE and MSI subtypes were also confirmed, respectively, by Howitt et al. and Pakish et al. [76]. Additionally, a study of 132 microsatellite stable, FIGO grade 2 endometrioid carcinoma patients [77] found that a subset of MSS ECs had higher expression of PD-L1. This study illustrated that among MSS tumors 48% (63/132) were PD-L1 positive and 16% (21/132) had more diffuse and/or especially strong PD-L1 expression. This PD-L1 over-expressed MSS cohort was associated with high numbers of tumor-associated CD3+ and CD8+ lymphocytes, sharing this characteristic with MSI ECs. In 2019, ESMO published a systematic review [78] in which the relationship of MSI status and PD-L1 expression was evaluated in several types of cancers. For EC, patients with both MSI and PD-L1 positive conditions only accounted for 3.1% of total ECs. Among all cancers studied, the cohort with MSI-high in combination with PD-L1 positive status was also a small percentage. Vanderwalde et al. [79] evaluated 11,348 cases (matched with 2189 cases) of 23 cancer types. The overall rate of PD-L1 positive cases was 25.4%, and only 26% of MSI-H cases were PD-L1 positive. Detecting the expression of PD-L1 by immunohistochemistry has been approved as a companion diagnostic test for the use of pembrolizumab in non-small-cell lung cancer (NSCLC), gastric/gastroesophageal junction adenocarcinoma, cervical cancer (CC), and urothelial cancer by the Food and Drug Administration (FDA) [80,81,82,83,84]. Furthermore, PD-L1’s predictive capacity has been seen across several other cancer types including head and neck and small-cell lung carcinoma [85,86,87]. Recently, for NSCLC patients with ≥50% PD-L1 expression on tumors, pembrolizumab was approved as a first-line therapy demonstrating a median overall survival (OS) of 30.2 months compared with 14.2 months for chemotherapy group. Based on these data, PD-L1 is a promising biomarker for immunotherapy in EC. However, research evaluating response to ICB based on the expression levels of PD-L1 has been inconsistent, and limitations still exist [88]. The heterogeneous expression of PD-L1 in the tumor microenvironment [88], lack of standard definition for positive-level of PD-L1 expression, and different detection methods resulting in lack of standardization across PD-L1 platforms are all problems which remain to be resolved.

Tumor mutational burden(TMB)

The second promising biomarker is TMB, defined as the number of mutations per coding region within the tumor genome [89]. In initial studies, mutation load was detected by whole exome sequencing comparing tumor DNA and matched normal DNA [90]. This expensive method is not widely applied in routine clinical care and now next-generation sequencing of targeted gene panels is commonly utilized to define TMB in clinical oncology [91, 92]. When normal cells undergo malignant transformation, cellular processes responsible for maintaining genomic integrity may be destroyed [93]. Cells are then unable to recognize or repair defects in DNA sequence or chromosome structure, resulting in accumulated mutations. Thus, it is no surprise that POLE-mutant and MSI ECs have a high TMB due to impaired DNA replication fidelity (POLE) and defective DNA MMR system (MSI-H) [14, 76]. DNA somatic mutations accumulate in cells, a fraction of which will further give rise to neoantigens [94]. These mutation-derived antigens can be displayed on HLA molecules on the surface of tumor cells [95, 96] and then be recognized by the immune system, especially after using T cell activating therapies [97,98,99,100,101]. Therefore, it is hypothesized that with higher TMB the ICB induced immune responses will be greater. Snyder et al. [102] first proposed TMB as a predictor of increased survival for patients who received ipilimumab or tremelimumab in melanoma. Subsequent trials defined TMB’s role across a wide variety of cancer types. For example, higher TMB was, found to benefit ICB therapies in NSCLC and small-cell lung cancer (SCLC) [103]. Currently, the FDA is considering approval of TMB-based assays as companion diagnostics for using ICB agents. As for MMR-d solid tumors which are defined as having a high TMB, recent work has demonstrated a high objective response rate (ORR) 53% to anti PD-1 therapy [104, 105]. These trials suggest the suitability of utilizing ICBs in TMB-high subtypes of ECs, which are mostly POLE and MSI ECs. Furthermore, although most cases of MSI-H solid tumors also have a high TMB, only 16% of TMB-high cases are MSI-H [91], suggesting that suitable cases for ICBs exist in other EC subtypes. What’s more, in addition to ICB therapy, TMB has shown predictive value in other immunotherapy modalities. Lauss et al. claimed that higher tumor mutation and neoantigen load could produce better clinical response (improved PFS and OS) in melanoma patients who were treated with adoptive T cell transfer therapy [106]. However, TMB also has some limitations as a biomarker. Some mutations in tumor cells can result in inactivation of the antigen presentation pathway causing no up-regulation in immune response [107, 108], impairing the predictive effects of TMB. Furthermore, there is no universal definition for high TMB [90]. Recent studies also evaluate the combination use of TMB with PD-L1 as biomarkers. Although the predictive roles of PD-L1 and TMB were shown to be independent, not correlated [109,110,111], and not co-associate in multiple trials [109], some trials did show a greater benefit of utilizing single anti-PD-1 and anti-PD-L1 therapy in patients with high TMB and PD-L1 expression [112]. Carbone et al. [109] showed that stage IV or recurrent NSCLC patients with both high tumor-mutation burden and ≥50% PD-L1 expression level had a better response to nivolumab than those with only one or neither of these biomarkers. The data suggest potential clinical value of combining TMB and PD-L1 as biomarkers for immunotherapy.

TIL

Another biomarker, tumor-infiltrating lymphocytes (TILs), was initially proposed by Clark at el. [113] to induce the lymphocytes in direct contact with tumor cells and/or infiltrating tumor nests. TIL includes a heterogeneous group of lymphocytes including effector T cells, tolerogenic or Treg cells, functionally exhausted T cells, NK cells, macrophages, DCs, MDSCs, and other immune cells [114]. These cell types have different competence in anti-tumor action. As the effector cells in TILs can elicit a cytotoxic antitumor immune response, the presence of TILs predicts better outcomes in several kinds of cancers such as melanoma [115], esophageal cancer [116], breast cancer (BC) [117] , colorectal cancer [118] and ovarian cancer (OC) [119, 120]. The predictive role of TILs was also demonstrated in EC by de Jong et al, who found high numbers of CTLs and high CD8+/ FOXP3+ ratio were associated with a longer disease free survival (DFS) while presence of CD45R0+ memory cells and high levels of CTLs predicted a greater OS [121]. As checkpoint inhibitors restore tumor cell recognition and T cell priming, pre-existing intratumoral CD8+T cells can exert anti-tumor response [122], suggesting the predictive role of TILs in immunotherapy. It has been found that patients with high “immunoscore”, representing the amount of infiltrating T cells, have better response to immunotherapies [123,124,125]. Because synthesized neoantigens due to accumulated DNA mutations are good binding sites for CD8+T cells, it makes sense that tumors with high TMB, such as POLE and MSI ECs, are the subtypes with higher TILs and benefit the most from immunotherapy [126, 127]. The TransPORTEC consortium performed assays on 116 high-risk ECs and confirmed that higher numbers of tumor-infiltrating T cells were found in POLE-mutant and MSI-H groups [128]. Recently, several other studies also showed increased TILs in POLE and MSI tumors compared with MSS tumors in EC and colorectal cancer [129, 130]. In 2017, Pakish et al. [76] compared the EC TME between the MSI-H group and MSS group (POLE-mutant or POLE-unknown cases were excluded) and reported increased immune cells, including granzyme B+ cells and activated cytotoxic T lymphocytes (CTLs, CD8+granzyme B+), were present in the stroma of MSI-H EC compared to MSS EC. Interestingly, this study further showed that Lynch syndrome (LS) related MSI-H ECs had reduced macrophages, increased CD8+ cells and activated CTLs compared with sporadic MSI-H ECs, suggesting the immune responses were affected by mechanisms underlying microsatellite instability.

Other emerging biomarkers

IDO1 is a type of tryptophan catabolic enzyme [131] which inactivates T cells and induces tumor immunotolerance [132]. IDO1 is over-expressed in a variety of tumor cells including EC cells [133, 134] and immune cells such as APCs, MDSCs and macrophages [132]. IDO1 expression (>25% ID O-positive) is more common in MMR-d EC (35%) than MMR-p EC (5%), specifically those with LS [135]. There is also a correlation between PD-L1 and IDO expression, as most PD-L1 positive tumors also express IDO [135], suggesting synergistic prediction in immunotherapy response.

Cyclooxygenase-2 (COX-2) is a rate-limiting enzyme responsible for the conversion of arachidonic acid to prostaglandins [136]. The production of COX-2 is usually at low levels in normal tissues [137] and is elevated in inflammatory states or cancer development [138, 139]. COX-2 is involved in a variety of procedures related to carcinogenesis, including angiogenesis, tumor cell proliferation, invasion, apoptosis inhibition, and immune evasion through prostaglandin E2 [140,141,142,143,144]. High levels of prostaglandin E2 can suppress interferon-γ(IFN-γ) production [145], antigen presentation [146] and IL-12 bio-synthesis and receptor expression [145, 147] and inhibit CTL proliferation and activation [148]. It has been reported that COX-2 overexpression was found in a series of cancers including CC, BC, gastric cancer, hepatocellular cancer and non-small cell lung cancer [149,150,151] and correlated with poor prognosis [152, 153]. In EC, a higher positive rate of COX-2 is related to more frequent cervical or extrauterine involvement (60.8%, p=0.02), higher grades (Grade 1 vs Grade 2 vs Grade 3: 13.6% vs 41.7% vs 60.9%, p=0.005), poor differentiation, and deep myometrial invasion [154, 155]. Additionally, COX-2 expression has been found to be associated with shorter DFS (p=0.09) [155] and disease relapse (p=0.03, univariate analysis) [156]. As for immune evasion in EC, COX-2 expression is inversely correlated with the degrees of CD8+T cell infiltration [156, 157]. This suggests that COX-2 can be used as a potential biomarker for immunotherapy.

Glycodelin, also termed as PAEP, is a secreted glycoprotein isolated from endometrium, decidua, seminal plasma and amniotic fluid [158]. It plays a suppressive role in cancer immune response which is similar to its function in maternal immune tolerance [159, 160]. The immuno-modulatory effects of glycodelin involve a variety of immune cells including DCs, NK, macrophages, T and B cells [161]. Over-expression of glycodelin is thought to play a role in carcinogenesis including the promotion of angiogenesis, cell proliferation, differentiation, and invasion [161], and is found in multiple gynecological cancers, such as EC and OC [162]. The expression of the immunosuppressive isoform of Glycodelin, termed Glycodelin A, is related to poor outcomes (p=0.003) and is an independent predictor for patient survival (p=0.002) in EC [163]. Although it has been identified as an important biomarker for immuno-modulatory functions in a certain cancers [164, 165], there is no data about its practical use in predicting immunotherapy efficacy, which deserves further study.

Several immune-related genetic signatures should be further investigated as alternative biomarkers for immunotherapy in EC. The AT-rich interaction domain 1A (ARID1A) mutation has been reported to not only facilitate ICB therapy but also potentially predict the efficacy of ICB therapy [166, 167]. Deficient ARID1A results in decreased MMR protein [168], higher mutation frequency [169] and increased TIL [170] and PD-L1 expression [171]. In EC, ARID1A alteration was correlated with higher infiltration of six common immune cells including CD8+ T and CD4+ T cells, B cells, neutrophils, macrophages and DCs [167], supporting the view that deficient ARID1A might be a potential predictor for ICB efficacy in EC. For hypomutated EC types including CN-low and CN-high/serous-like ECs, recent studies noted the correlations of several gene mutations with low neoantigen load, such as CTNNB1 alteration in CN-low EC and MYC amplification and PIK3CA alteration in CN-high EC [14, 172]. These genetic biomarkers might predict poor effects of immunotherapy response and can be used to select potential candidates with non-immunogenic types of EC for immunotherapy [173].

ICBs

ICBs are a group of monoclonal antibodies targeting immune checkpoint proteins which mainly transmit co-inhibitory signals during T cell activation [174]. In the immune microenvironment of EC, tumor elicited immunosuppression is mainly generated from the conjugation of over-expressed PD-L1 and PD-L2 on EC cells to PD-1 receptors on tumor infiltrating CD4+/ CD8+ T cells and CTLA-4 expressed on Treg to B7-1/B7-2 (the ligand of stimulatory receptor CD28) expressed on APC. ICBs can decrease the negative immunomodulation exerted by tumor cells/Tregs through PD-1/PD-L1 and CTLA-4/B7-1, B7-2 pathways and thus restore antitumor effects of T cells [67, 175, 176]. Since 2011 when ipilimumab (anti-CTLA4) was first approved by the FDA for patients with metastatic melanoma, various ICBs, such as nivolumab, pembrolizumab, atezolizumab, avelumab and durvalumab, have continuously been approved by the FDA for treating a wide range of malignancies, with approval likely for additional tumor types in the near future. POLE and MSI ECs are favorable candidates for ICBs therapy because of high expression of immune-related biomarkers which have been previously illustrated. One of the ICBs, pembrolizumab (anti-PD-1 agent), has been approved by the FDA for patients with MSI or MMR-deficient solid tumors which are resistant to conventional treatment, supporting ICB therapy in the treatment of advanced MSI EC. Additionally, the expression of canonical biomarkers and novel evidence of ICB adaptability have been detected in a portion of EC patients aside from POLE/MSI types, such as the MSS EC.

ICBs in POLE/MSI EC

Pembrolizumab, a PD1 blockade, is the first ICBs whose clinical activity has been investigated in EC. A phase II trial (NCT01876511) by Le et al. [104] evaluated the efficacy of monotherapy pembrolizumab (10 mg/kg every 14 days) in patients with advanced MMR-deficient cancers (12 tumor types). This trial included 15 MSI EC patients. For the EC cohort, after trial completion, the disease control rate was 73% including 3 (20%) patients with complete response (CR), 5 (33%) patients with partial response (PR) and 3 (20%) patients with stable disease (SD). The ORR of the EC cohort was 53%. Treatment related adverse events (AEs) were observed in 74% of patients, most of which were at low grade and manageable, such as endocrine disorders (i.e. hypothyroidism). This trial expanded the therapeutic value of ICBs from canonical MMR-d colorectal cancers to more MMR-d tumor types and drew researchers’ attention to apply ICBs in MSI EC.

Pembrolizumab was then evaluated in a multicohort phase Ib KEYNOTE-028 (NCT02054806) study which enrolled patients with PD-L1 positive advanced solid tumors [177]. Twenty-four advanced PD-L1 positive EC patients were eligible, among whom, only 2 patients were defined as POLE and MSI-high respectively while others were non-MSI-high or not evaluable. Twenty-three EC patients were included in the final efficacy analysis, the ORR was 13.0% (95% CI, 2.8% to 33.6%), the median PFS at the data cutoff was 1.8 months (95% CI, 1.6 to 2.7 months), and the median OS was not reached (95% CI, 4.3 months to not reached). PR, SD, and progressive disease (PD) were observed respectively in 3 (13.0%), 3 (13.0%), and 13 (56.5%) patients. The therapy in the single POLE patient was particularly effective as he achieved PR,while the MSI-high patient had PD. Grade 3 treatment-related AEs only occurred in four patients and no grade 4 AEs or AE induced treatment discontinuations were observed. This study suggested a superior benefit of PD-1 blockade in POLE ECs, but the analysis was limited by small cohort sizes.

In 2019 a phase II KEYNOTE-158 study (NCT02628067) evaluating pembrolizumab in MMR-d noncolorectal carcinoma was published. This study enrolled a larger cohort of EC patients [178]. Forty-nine patients with progressive MSI EC, intolerant to standard therapy, were eligible. The median follow up time was 13.4 months. The total ORR for all 23 MMR-d tumor types was 34.3% (95% CI, 28.3% to 40.8%). For EC cohort, 8 (16%) patients achieved CR, 20 (41%) patients achieved PR, the ORR was 57.1% (95% CI, 42.2 to 71.2) , the median PFS was 25.7 (95% CI, 4.9 to NR), and the median OS was not reached. Specifically, EC was one of the tumor types with the most frequent CR in this study. Furthermore, 37 EC patients had a ≥30% reduction in tumor size among 47 EC patients with a tumor size change from the baseline data, and EC was the most common tumor with size reduction in the study. Severe AEs occurred at a low rate as only 3 patients experienced grade 4 treatment-related AEs and one patient died of treatment induced pneumonia were reported. These data show the impressive value of using pembrolizumab in MSI EC. Ongoing trials are investigating combination therapy of pembrolizumab with other therapeutic methods in MSI EC. A phase I/II study (NCT04014530) of MSI metastatic EC and colorectal adenocarcinoma are recruiting volunteers for pembrolizumab plus ataluren combination therapy with the primary outcome measure of ORR. Trials investigating combination of pembrolizumab with radiotherapy (NCT04214067) or anti-angiogenic agent (NCT04197219) in MSI EC have also been registered but not yet recruited yet.

Recently, nivolumab, a new PD-1 blockade, has shown great activity in MMR-d colon cancer. Mounting preclinical or clinical studies are ongoing to broaden its usage in more MMR-d tumors. In 2020, a newly published study noted that the effect of nivolumab was evaluated in a phase II study (NCT02465060) which enrolled 42 relapsed MMR-d non-colorectal cancer patients [179]. Thirteen refractory/relapsed MSI EC patients were eligible. Nivolumab was administrated intravenously 3 mg/kg every 2 weeks (28-day cycles) and 480 mg every week after cycle 4. The total ORR was 36% (15/42; 90% CI,23.5% to 49.5%). Three patients (7%) achieved CRs, 13 patients (29%) achieved PR and 9 patients (21%) achieved SD. As for the EC cohort, 2 patients had CR (2/13, 15%), 3 patients had PR (3/13, 53%) and 7 patients showed decreased tumor size from baseline (7/13,53%). Treatment-related toxicities were mild and at low grades (most were grade 1-3), with only 2 grade 4 AEs in 3 patients and no grade 5 AEs. This trial noted a promising effect of nivolumab on treating MSI EC. An ongoing phase 2 study (NCT03241745) of nivolumab monotherapy is recruiting patients with MSI metastatic /recurrent uterine cancer, which will further provide evidence on using nivolumab in MSI EC.

Blocking PD-L1 is another significant mechanism for ICBs in EC. In 2017, the effect of a PD-L1 blockade, atezolizumab, was initially evaluated by a phase Ia study (NCT01375842) in 15 advanced or recurrent endometrial cancer (rEC) patients [180]. Among them, 1 was MSI-H, 7 were MSS, and 7 had unknown MMR status. Atezolizumab 1200 mg or 15 mg/kg intravenous every 3 weeks was administered to the patients. PD-L1 expression was evaluated in the tumor samples with expression on immune cells (IC) comprising ≥ 5% of the tumor defined as PD-L1 positive (IC2/3). After treatment, the ORR was 13% (2/15) in total and 40% (2/5) in IC2/3 patients. As for the 2 responders who achieved PR, one was MSI-H and had moderate TILs infiltration (IC2,10%TILs), the other was MSS but had heavy TILs infiltration (IC3,70%TILs) which was similar to the tumor immune micro-environment in MSI ECs. Duration of objective response (DOR) for these 2 cases were 7.3 and 8.1+ months, respectively. Median PFS was 1.7 months (range, 0.6-11+); median OS was 9.6 months (range, 0.6-11.8+). Atezolizumab showed a relatively safe profile in rEC. Among 7 (47%) patients with any treatment related AEs, 5 patients had G1-2 AEs, no G4-5 AEs was observed, and only 2 patients had SAEs including colitis and rash. This trial noted that MSI status, high TILs, and PD-L1 positivity may be factors for atezolizumab therapy.

Aside for atezolizumab, avelumab is also a promising immune check point inhibitor targeting PD-L1 in EC. Konstantinopoulos et al. [181] performed a phase II study (NCT02912572) of avelumab in patients with recurrent/persistent EC. Thirty-three patients were enrolled in this study and were divided into 2 cohorts: MMR-d (17/33) and MMR-proficient (16/33), with no POLE patients enrolled. Avelumab 10 mg/kg was administered intravenously every 2 weeks until tumor progression or unacceptable toxicity. Two MMR-d patients were not enrolled in protocol treatment and were excluded from terminal analyses. At the first stage, only 1 patient from the MMR-p cohort achieved both objective response (OR) and PFS at 6 months (PFS6), so this cohort was closed because of futility and did not enter into next stage, the ORR was 6.25% (95% CI 0.16% to 30.2%). The MMR-d cohort completed 2 stages of treatment. In this cohort, the ORR was 26.7% (95%CI, 7.8% to 55.1%), 4 ORs including 1 CR and 3 PRs were observed. Six patients exhibited PFS6 (40.0%; 95% CI 16.3% to 66.7%, including all 4 ORs), 4 of whom were still receiving the treatment at the data cutoff. Treatment-related toxicity was tolerable. All treatment-related AEs were G1-3, with no G4 or G5 AEs occurring in either cohort. This study suggested promising effect of avelumab in MMR-d EC.

Durvalumab, another PD-L1 inhibitor, also showed impressive effects in MSI ECs. In 2019, 71 patients with advanced EC were enrolled in a phase II PHAEDRA trial (ANZGOG1601) [182] to evaluate the effect of durvalumab therapy. These patients had all experienced progression after 0-3 lines of chemotherapy prior to the durvalumab treatment. Thirty-six patients were defined as MMR-d and 35 patients were MMR-p. Durvalumab was administrated 1500mg intravenous Q4W. The objective tumor response (OTR, including CR or PR by Response Evaluation Criteria In Solid Tumors) rate for MMR-d cohort was 47% (17/36, 95% CI 32-63%), including 6 CR and 11 PR, median PFS was 5.5, 12-month OS was 71% and median OS was not reached. In contrast,for the MMR-p cohort, the OTR rate was only 3% (1/35,95% CI 1-15%), 1 PR and 10 SD were observed, median PFS was 1.8 months, 12-month OS was 51%, and median OS was 11.5 months.

As for CTLA-4 targeted therapy, data are limited in POLE/MSI EC. Ipilimumab and tremelimumab are two significant anti-CTLA-4 monoclonal antibodies in the clinical studies of melanoma, mesothelioma, NSCLC [183,184,185] and other tumors. Several trials of CTLA-4 combination therapy in EC are ongoing to identify the possible clinical efficacy. Rubinstein et al. [186] reported an interim analysis of a phase II trial (NCT03015129) comparing the combination of durvalumab and tremelimumab (DT arm) to durvalumab monotherapy (D arm) in patients with advanced EC and endometrial carcinosarcoma. Fifty-six patients were enrolled and divided equally into D arm and DT arm, 5 (9%) patients were MSI-H, 48 (86%) patients were MSS and 3 (5%) patients had unknown MMR status. Twenty-seven patients per arm were enrolled in the evaluation and a modest clinical activity was observed. 40% (2/5) of MSI patients had ORs (D arm: 1 CR, DT arm: 1 PR). The efficacy appeared poorer in MSS patients with only 5 (10.4%) achieving ORs (D arm: 1 CR,2 PR, DT arm: 1 CR, 1 PR). In this trial, G3 and G4 treatment-related AEs were observed in 11 (DT arm: 9, D arm: 2) and 4 (DT arm: 3, D arm: 1) patients, respectively. However, serious AE induced by this kind of combination strategy was described in a case report by Mahmood et al [187]. They reported on a 75-year-old Caucasian female with advanced EC received durvalumab 1500 mg plus tremelimumab 75 mg as a combination therapy and only had grade 1 skin pruritus at the first cycle of treatment. However, after 4 weeks, she developed a fulminant immune-mediated myocarditis and was treated with high-dose intravenous steroid. She did experience cardiac function recovery but had progression of metastatic lesions. Recently, a phase II study (NCT02982486) began investigating the combination of ipilimumab (an anti-CTLA-4 agent) and nivolumab in patients with nonresectable/metastatic sarcoma or EC with somatic MMR deficiency. This trial will evaluate the efficacy of ipilimumab 1 mg/kg every 6 weeks plus nivolumab 240 mg every 2 weeks in an estimated 60 participants, and CR and PR will be the primary outcome measures.

ICBs in MSS EC

Although POLE/MSI ECs have shown considerable sensitivity to ICB therapy, these two subtypes only comprise a minority portion of EC cases. Most of the endometrioid (72%) and serous (98%) EC patients fall into the copy number-low or copy number-high subtypes, with no MSI characteristics [188]. Sporadic responses of non-POLE/MSI ECs were seen in the trial results listed in the prior paragraphs. Furthermore, Goodman et al. [189] claimed that patients with MSS tumors but marked as TMB-high might benefit from immunotherapy, suggesting complicated factors contributing to clinical outcomes aside from the known biomarkers.

In March 2019, Makker and his colleagues [190] published an interim analysis of a phase 2 trial (NCT03015129) which assessed the combination of pembrolizumab and lenvatinib in advanced endometrial cancer. Eligible patients were unselected for microsatellite instability or PD-L1 expression status. At the interim analysis cutoff, 53 patients were included in this interim evaluation, 85% (45/53) of whom were defined as MSS and 25% were PD-L1-positive. At 24 weeks, 39.6% (21/53) of patients had an OR. The median follow-up for PFS was 7.7 months. Notably, objective responses and tumor size reduction from baseline were respectively observed in 35.6% (16/45) and 80% (36/45) of MSS patients. This was higher than what was reported in former studies of advanced EC [191, 192]. This result led to the approval of pembrolizumab plus lenvatinib by the FDA for the treatment of advanced EC, a significant breakthrough. In October 2019, the final result of the advanced EC cohort in this trial was published. At data cutoff, 108 patients were included in the final analysis, MSI vs MSS was 13% (11/108) vs 87% (94/108), 38% (41/108) of total patients reached ORR at 24 weeks (95% CI, 28.8-47.8%), the median duration of response was 21.2 (7.6-NR) months. The ORR at 24 weeks was 36.2% (34/94) vs 63.6% (7/11) for MSS vs MSI, 7 (7.4%) MSS patients and 1 (9.1%) MSI patient achieved CR, and 28 (29.8%) MSS patients and 6 (54.5%) MSI patients achieved PR. The median duration of response was not reached for the MSS cohort and was 21.2 months for the MSI cohort. In conclusion, the authors reported promising activity of pembrolizumab plus lenvatinib in advanced EC regardless of MSI/MMR status. Two phase 3 studies (NCT03517449, NCT03884101) are ongoing to further examine lenvatinib plus pembrolizumab versus chemotherapy regimens of doxorubicin/paclitaxel/carboplatin in advanced EC with known MMR status, in order to provide more data to guide the use of this therapy.

There are also some case reports discussing the latent benefit of using ICBs in MSS EC. For example, in 2019, Oh and Chae [193] reported a 57-year-old, MMR-proficient EC patient with diagnosis of stage IV endometrial adenocarcinoma and PD-L1 negative status. After completion of neoadjuvant chemotherapy, total abdominal hysterectomy and bilateral salpingo-oophorectomy with optimal surgical debulking and 3 cycles of chemotherapy, she had PD at 4 months and wanted to avoid continued chemotherapy. She was then introduced to a combination PD-1 and CTLA-4 blockade, which included nivolumab 3 mg/kg every 2 weeks and ipilimumab 1 mg/kg every 6 weeks. Fortunately, she had a deep and durable response after this treatment with 79% shrinkage of tumor size in 1-year, continued reduction of metastatic lesions, and PR noted by cross-sectional imaging. This case report highlights a satisfactory clinical response to ICB in an MSS EC patient who would typically have been considered an unfavorable candidate for immunotherapy.

Although clinical responses are sometimes seen in MSS EC, conclusive biomarkers recognizing responders are still lacking [188]. Current studies on treating MSS EC by immunotherapy mainly concentrate on combining ICBs with other agents to get a higher anti-tumor effect as described before. There exists a series of ongoing trials studying the combination of ICBs with chemotherapy, radiotherapy, poly (ADP-ribose) polymerase (PARP) inhibitors, antiangiogenic drugs, and other target agents. Patients enrolled in these assays will be evaluated for MMR. The prospective outcomes of patients with diverse MMR status in those trials will enlighten us on the subject of immunotherapy in MSS EC patients. Representative trials are listed as follows.

ICBs & Antibody-drug conjugates(ADCs)

ADCs are a group of agents composed of a specific antibody targeting tumor-associated antigen conjugated with a cytotoxic effector compound [194, 195], which have been identified as effective therapies in solid tumors [196, 197]. Mirvetuximab soravtansine (IMGN853), as an ADC, involves a humanized anti-FRα monoclonal antibody targeting selective tumor cells and tubulin-disrupting maytansinoid DM4 as a cytotoxic module [195, 198]. After the binding of IMGN853 to FRα on tumor cells, the drug is internalized, leading to an accumulated intracellular concentration of DM4 [199]. This produces an antimitotic effects, and the tumor cells are killed [200, 201]. The combination of IMGN853 with pembrolizumab is being investigated in patients with MSS recurrent or persistent EC in an ongoing phase 2 study (NCT03835819). Thirty-five participants are estimated to be enrolled and will receive both pembrolizumab and IMGN853 administered intravenously once every 3 weeks. The primary outcome measures include ORR and PFS.

ICB & antiangiogenic agents

Aside from the previously described trials of pembrolizumab plus lenvatinib, several ongoing trials are researching the combination therapy of ICBs with antiangiogenic agents. There is a phase II, single arm study (NCT03526432) investigating atezolizumab plus bevacizumab which is now recruiting patients with advanced EC. Patients’ MMR status will be determined before they enter into the trial. PR and CR will be measured for the evaluation of clinical efficacy as well as PFS, OS, and number of patients experiencing toxicity and immune related response.

ICB & PARPi

A multicenter, placebo-controlled, phase III study (NCT04269200) is ongoing to investigate the combination therapy of first-line chemotherapy, durvalumab, and PARPi in patients with newly diagnosed advanced EC. Six-hundred-ninety-nine EC patients with known MMR status are estimated to be enrolled in this trial. This study includes 3 arms: patients in arm A receive standard chemotherapy of carboplatin and paclitaxel and placebos as the control group, patients in arm B receive chemotherapy plus durvalumab, and the residual patients in arm C receive the combination therapy of chemotherapy, durvalumab, and olaparib. The primary outcome measure is PFS. Since ARID1A gene deficiency in some EC patients is related to DNA homologous recombination repair deficiency [202], PARPi may be effective in these ARID1A mutated patients, which is supported by a series of preclinical trials [203, 204].

ICBs & IDO1 inhibitors

BMS-986205(69,(R)-N-(4-chlorophenyl)-2-((1S,4S)-4-(6-fluoroquinolin-4-yl)cyclohexyl)propanamide) is an IDO1 inhibitor with potential [132] therapeutic benefit, as it has been demonstrated that IDO1 is partly responsible for the formation of resistance to ICBs [131]. Combination of IDO1 inhibitor (such as BMS-986205) with ICBs might be an alternative strategy for EC patients. The combination of nivolumab with BMS-986205 in recurrent or persistent EC and endometrial carcinosarcoma is being investigated in an ongoing phase 2 trial (NCT04106414). Enrolled patients will be divided into a nivolumab monotherapy (480 mg every 4 weeks) group and a nivolumab (480 mg every 4 weeks) plus BMS-986205 (100 mg every 4 weeks) group. MSI/MSS evaluation must be done prior to entering into the trial. The best overall response rate determined by RECIST 1.1 is the primary outcome measure.

Emerging immune checkpoints and ICBs

Lymphocyte-activation gene 3 (LAG-3, CD233)

New ICBs are being researched to provide a large selection of therapeutic drugs. LAG-3 is a key immune inhibitory receptor mainly expressed on activated T and NK cells [205]. Previous studies identified the major histocompatibility complex class II (MHC-II) as the major ligand of LAG-3 [206, 207]. The combination of LAG-3 with MHC-II can inhibit the activation of CD4 + helper T cells which depend on MHC-II/CD4 interaction [208]. However, this mechanism fails to explain the functional suppression of CD8+ T cells and NK cells by LAG-3 [209] or the T cell activation by several anti-LAG3 mAbs which fail to target LAG-3/MHC-II signaling [210,211,212,213]. In January 2020, Wang et al. [213] suggested fibrinogen-like protein 1, which is highly produced by cancer cells, as the major ligand for LAG-3. The fibrinogen-like protein 1/LAG3 combination could induce LAG3-dependent T cell suppression and result in tumor immune evasion. These findings support LAG-3 as an important target for ICB development. A phase 1 study (NCT03538028) of INCAGN02385 (an antagonist antibody targeting LAG-3) in patients with advanced tumors including MSI EC is now in the recruiting period, and the primary outcome measured will be the number of treatment-emergent adverse events (TEAEs).

B7-H4

B7-H4 is a coinhibitory molecule contributing to the B7 family [214, 215]. It binds to unknown receptors on activated T cells [216] and transmits negative immuno-modulatory signals, thus promoting immune escape through negative regulations of cytokine secretion, cell proliferation, and the T cell cycle [217]. B7-H4 is highly expressed on tumor-associated macrophages [218] and in many solid tumors, such as OC, BC, lung cancer, renal cell cancer, and pancreatic cancer [219,220,221,222,223,224]. Different results were observed in studies testing the expression levels of B7-H4 in EC. Miyatake et al. [225] found that with progression of the endometrial mucosa from a normal phenotype to hyperplasia and malignancy, a higher proportion and intensity of B7-H4 expression was observed on the surface of the cells. Additionally, high risk ECs had a higher intensity and proportion of B7-H4 positivity compared with low risk ones ( P=0.001 and P=0.032, respectively), and B7-H4 was positively associated with CD3+ and CD8+ TILs ( P=0.039 and P=0.031, respectively). However, Vanderstraeten et al. [134] and Liu et al. [226] found a high rate of B7-H4 positivity (90-100%) in EC tissues regardless of the cancer settings (primary or metastatic) and the normal endometrial samples in these two studies were all (100%) B7-H4 positive. Also, Bregar et al [227] claimed that the expression levels of B7-H4 were similar among ECs with different microsatellite status, grades, histology determinations, and immune cell infiltrations. Recently, the role of B7-H4 in tumor development has been identified. It is tightly associated with tumor aggressiveness and metastasis by functioning to promote cancer cell proliferation, invasion, and anti-apoptosis [217]. Therefore, B7-H4 has been considered a novel target for immunotherapy in cancer. An ongoing (NCT03514121) phase Ia/b trial is now testing FPA150 (an anti-B7-H4 antibody) in patients with advanced or metastatic solid tumors including EC. This study will evaluate the maximum tolerated dose (MTD) and/or recommended dose (RD), and AEs will be recorded.

T cell immunoglobulin and mucin-domain containing-3 (TIM-3)

TIM-3 is another intriguing immune checkpoint molecule which is similar to PD-1 or CTLA-4 [228]. TIM-3 is not only expressed on effector T cells (CD4+ and CD8+ T cells) but is also found on Tregs, B cells, NK cells, and antigen-presenting cells including DCs and macrophages [229]. TIM-3 can induce the exhaustion of cytotoxic T cells and activation of Tregs resulting in immune-tolerance via a mechanism separate from the PD-1/PD-L1 axis [230,231,232,233,234]. The expression of TIM-3 has been identified in a variety of solid tumors, such as cervical, urothelial, gastric, and prostatic carcinomas and melanomas [234,235,236,237,238]. As for EC, Moore et al. [239] demonstrated that focal expression of TIM-3 was found in all EC cases, but stronger expression was observed in MMR-d cases (66% of MMR-d vs 12% of MMR-p, P=0.00002) with particularly intense TIM-3 staining in MLH1-hypermethylated and MSH6 loss cases. Furthermore, intermediate and high-grade EC are more likely to express TIM-3 compared with low grade tumors (P=0.02). Although this immune checkpoint has shown potential value as a target for ICB therapy in EC, no related trials or research exist, so the clinical usage of TIM-3 in EC remains to be explored.

Cancer vaccine

A cancer vaccine is a form of active-specific immunotherapy (ASI) [240] which harnesses the host’s immune system to attack the tumor cells [241]. The rationale for designing cancer vaccines is similar to that of vaccines against infectious diseases [240], which can be summarized as employing disease/tumor associated specific antigens to elicit APC mediated CD8+ and CD4+ T cell responses [242] and induce a persistent immune memory [243]. Under the tumor elicited immuno-tolerant environment [244], high levels of cancer vaccine are necessary for the expansion of DCs [245], which in turn promote the anti-tumor effects of T cells. Cancer vaccines are further classified as prophylactic vaccines and therapeutic vaccines [246]. The prophylactic vaccines, such as the HPV and HBV vaccines, block the infection of tumor-driving viruses and are only preventive if its administration precede tumor occurrence [246]. There is not the main topic discussed here. The categories of therapeutic cancer vaccines mainly include tumor/immune cell vaccines, peptide vaccines, and genetic vaccines comprised of DNA, RNA, and viral vaccines [247].

Peptide vaccine

Specific tumor associated antigens over-expressed on EC cells can be artificially processed to generate peptide cancer vaccines. One example of such is the WT1 peptide vaccine. The wild-type WT1 gene has been characterized as an oncogene in many malignancies [248] and is specifically highly expressed in various gynecological cancers [249, 250]. The product of the WT1 gene is a tumor associated antigen, which can be recognized by T cells when associated with MHC class 1 molecules. Since 2004, a number of preclinical studies and case reports of WT1 immunotherapy have been published [251, 252]. Direct injection of modified WT1 peptide as a vaccine can induce a WT1-specific immune response [253]. OHNO et al. [254] performed a phase II clinical trial investigating a WT1 vaccine in patients with WT1/HLA-A 2402-positive gynecological cancers. Twelve enrolled patients were given 3.0 mg of HLA-A 2402-restricted, modified 9-mer WT1 peptide every week for 12 consecutive weeks. During the 3 months trial period, the disease control rate was 25.0%; however, no cases experienced a CR or PR with this therapy. To explore a more effective vaccination, administration of autologous DCs loaded with WT1 peptides gained attention. In 2013, Coosemans et al. [253] published a report of a Phase I/II Trial (EudraCT 2009-016868-37) investigating WT1-loaded dendritic cell immunotherapy in patients with uterine tumors. Among the 6 enrolled patients, 3 were diagnosed with serous endometrial carcinoma. Four of the patients were HLA-A2-positive including 2 EC patients. All patients were given autologous DCs transfected with WT1 mRNA as vaccines weekly for 4 weeks, and 2 HLA-A2-positive patients received 2-3 additional injections. As a result, 75% (3/4) of HLA-A2-positive patients showed an immunological response after four injections including the 2 HLA-A2-positive EC patients who also demonstrated increased WT1-specific T-cells and NK cells. No oncological or immunological response was observed in the 2 HLA-A2-negative patients which included 1 EC patient.

As granulocyte-macrophage colony-stimulating factor (GM-CSF) has been found as the most valid anti-tumor cytokine through systematic selection on cytokine panels in murine models [255], combining cancer vaccines with GM-CSF might have a synergistic anti-tumor effect. Folate-binding protein is highly expressed in various malignant tumors [256, 257]. E39 is an immunogenic, HLA-A2-restricted peptide derived from folate-binding protein and is the most consistent recognition site of tumor-associated lymphocytes. In a phase I/IIa trial (NCT01580696) [258], the clinical activity of E39 peptide vaccine in combination with GM-CSF was explored in ovarian and endometrial cancer patients. Fifty-one enrolled patients were divided into 2 groups: 1. vaccine group: HLA-A2 positive cases receiving E39+GM-CSF vaccines (29 cases) and 2. control group: HLA-A2 negative cases or HLA-A2 positive cases rejecting vaccines (22 cases). Nine EC patients were enrolled in this trial: 6 in the vaccine group and 3 in the control group. The 24-month DFS was 55.5% for all vaccine group patients and 40.0% for all control group patients (P= 0.339). The 24-month DFS of EC patients in vaccine group was 62.5%, which was higher than that of total vaccine group. The vaccine was well tolerated with no greater than G2 local toxicities and no greater than G3 systemic toxicities observed.

NY-ESO-1 is a cancer/testis antigen which expressed both on testis and various human malignancies [259,260,261]. Jäger et al. [262] conducted a trial to evaluate the safety and immunogenicity of using NY-ESO-1 vaccine in patients with 8 kinds of advanced NY-ESO-1-expressing tumors. Thirty-six enrolled patients were given recombinant vaccinia-NY-ESO-1 (rV-NY-ESO-1) and a fowlpox-based vaccine containing NY-ESO-1. Twenty-three patients finished four vaccinations and entered the final analysis for tumor and immunological response. NY-ESO-1-specific T cell and antibody responses were observed in the majority of cases. The different patterns of immune responses observed in patients after treatment were divided into four categories, from the lowest category I to the highest category IV, based on serologic conversion and T cell response. The single stage IV EC patient was classified into Category III, which included converting from sero-negative to sero-positive and improved CD4+ and/or CD8+ T cell responses. Although she showed obvious immunological response, she had progressive disease.

In addition to tumor associated antigens, epitopes of immune cells are good candidates for manipulating vaccines. A phase I trial in patients with metastatic malignancies published in 2009 by Kaumaya et al. [263] evaluated a chimeric peptide vaccine which combined B-cell epitopes derived from the human epidermal growth factor receptor 2 (HER2) extracellular domain with a promiscuous T cell epitope. This combination vaccine was emulsified in Mon-tanide ISA 720 (SEPPIC, Inc., Paris, France) with nor-muramyl-dipeptide (n-MDP) adjuvant. Twenty-four patients received 3 vaccinations and 25% (6/24) exhibited clinical benefit. Two EC patients were included in the final analysis with only 1 having a clinical PR. This study demonstrated that the chimeric vaccine was safe as no greater than G3 treatment related AES were observed. This type of combination vaccine was further studied, and a recent publication in 2019 of a phase I trial (NCT01417546) showed antitumor activity with this vaccine type in patients with advanced solid tumors in a phase I trial, but no EC patients were included [264].

Nucleic acid-based vaccines

mRNA-4157 is a personalized vaccine designed by a proprietary algorithm. It is a lipid encapsulated personalized vaccine which elicits specific anti-tumor T cell responses through the vaccine encoding neoantigens. A phase 1 Keynote-603 study (NCT03313778) is ongoing to evaluate the safety and immunogenicity of mRNA-4157 monotherapy in patients with resected solid tumors or mRNA-4157 plus pembrolizumab in patients with unresectable solid tumors. In the monotherapy group, mRNA-4157 was given 0.04 - 1 mg every 3 weeks for 9 cycles; in the combination group, patients first received 2 cycles of 200 mg pembrolizumab, then received mRNA-4157 plus pembrolizumab for 9 cycles, which may be followed by a pembrolizumab monotherapy for up to 2 years. In May 2019, an interim report of this trial was published and up to 33 patients were enrolled in the treatment. One MSI-high EC patient was included in the combination therapy cohort, with the remaining tumor types including bladder , HNSCC, melanoma, NSCLC, SCLC, MSI-high CRC, MSI-high prostate and TMB-high metastatic cutaneous squamous cell. Although the outcome of the only EC patient was not provided, the clinical responses for the whole combination therapy cohort, including 5 PRs, 6 SD, and 8 PD, were noted with no SAEs or AEs ≥ G3 supporting the advancement of mRNA-4157 to future trials.

In MSI EC cells, MMR deficiency can result in length changes of microsatellite sequences within coding regions of the human genome, which is termed as coding MSI [265]. Coding MSI can generate frameshift peptides (FSPs) which can promote oncogenesis through functional inactivation of tumor-suppressive proteins and induce tumor-specific immune responses [266]. FSPs are not found on normal human cells, and the expression patterns of MSI-related FSPs between sporadic MSI tumors and hereditary MSI tumors show no difference. Thus, based on the immunogenicity and consistent expression in MSI tumors, MSI-related FSPs have become candidates for developing cancer vaccines in MSI EC. In 2019, a phase I, First In Humans, multicenter study of Nous-209 genetic polyvalent vaccine combined with pembrolizumab in patients with MSI solid tumors was posted and is currently recruiting patients. Nous-209 Genetic Vaccine is a heterologous prime/boost regimen composed of GAd20-209-FSP (priming) and MVA-209-FSP (boosting), which encodes 209 distinct FSP cancer neoantigens found in various MSI tumors including MSI EC. Thirty-four patients are estimated to be enrolled and will be administrated 1 priming dose of GAd20-209-FSP followed by 3 boots with MVA-209-FSP in combination with pembrolizumab. Dose limited toxicity and Treatment-Emergent AEs will be evaluated for this regimen as primary outcome measures.

ACT

ACT, a passive immunotherapy, means isolating allogenic or autologous immune cells, which are activated and expanded ex vivo followed by reinfusion into cancer patients [267]. This form of immunotherapy has several advantages, including production of multiple tumor-specific lymphocytes in vitro, alternative engineered immune cells for specific tumor antigens and favorable host environment due to lymphodepletion prior to ACT [268]. CTL and NK cell are principal effector cell types used in ACT, while DC are usually used as a tool to carry vaccine or to present antigens to stimulate T cells in vitro [268, 269]. Additionally, transforming genetically modified T or NK cells (such as CAR T or CAR NK cells) are emerging strategies in ACT and have shown activity in a majority of malignancies. However, ACT also has several limitations: short-period effects and poor trafficking of effective cells within immunosuppressive environment [268]. Current studies related to ACT in EC involve transferring Lymphokine-activated killer (LAK) cells, common DC primed T cells, CAR T cell therapy and other complex combination therapies.

Adoptive LAK cells transfer

LAK cells is a group of lymphocytes mainly comprised of T and NK cells which are generated by patient derived peripheral blood mononuclear cells exposed to high dose interleukin-2 (IL-2) [270,271,272]. The preclinical study of LAK cells in EC was first published in 1989. Shimizu et al. [273] proposed that the combination of adoptive transfer of LAK cells in combination with intraperitoneal injection of recombinant IL-2 (rIL-2) markedly inhibited the growth of human EC cell line xenografts in nude mice. The adoptive transfer of LAK cells plus IL-2 was further performed in clinical patients in a phase I trial by Steis et al. [274]. Twelve patients with colorectal cancer, 10 patients with OC, 1 patient with small-bowel adenocarcinoma, and 1patient with EC, all with malignancies limited to the peritoneal space, were enrolled in this trial. A modest therapeutic efficacy was observed as 30% of the patients had PR in this trial. However, the single one EC patient showed no responses. Additionally, this kind of ACT requires improvement as multiple and significant toxicity-related AEs were observed. All patients involved developed diffuse abdominal pain and rebound abdominal tenderness, which was assumed to be partly related to the injection of LAK cells. Treatment related intraperitoneal fibrosis was another severe complication of this strategy.

Adoptive T cell transfer

Adoptive T cell transfer is a significant component of ACT therapy. Santin et al. [275] investigated the adoptive transformation of DC primed peripheral blood T cells in a 65-year-old woman with unresectable and chemoresistant EC. The autologous DC were first treated with autologous tumor lysate and then were used to generate activated tumor-specific T cells. During the treatment, the patient showed stabilization of a liver metastasis which had markedly increased from 9.5 x 8 cm to 14 x 10 cm in the 3 weeks prior to the treatment. The patient showed a SD response during the treatment and maintained a stable status for at least 3 weeks after the final infusion.

T cells can be genetically modified through introducing chimeric antigen receptors (CAR) by retrovirus or a lentivirus [276]. CARs are comprised of 3 modules: an extracellular target binding module, a transmembrane module, and an intracellular module [277], which can provide both tumor-associated antigens recognition and T cell activation [278]. CAR T-cell therapy was a breakthrough in the treatment of lymphoid malignancies, such as Acute Lymphoblastic Leukemia (ALL) [279, 280] and Diffuse Large B-cell Lymphoma (DLBCL) [281]. CAR T-cell therapy related research results are still lagging behind in EC. Rodriguez-Garcia et al. [282] established a preclinical study of anti-Müllerian inhibiting substance type II receptor (MISIIR) CAR T cell therapy in patients with ovarian and endometrial cancer. The MISIIR, as a member of the transforming growth factor-β receptor family, is highly expressed on the majority of gynecologic cancers [283,284,285,286,287,288], making it an ideal target for CAR T cell therapy. The anti-tumor activity of anti-MISIIR CAR T cell therapy in EC was examined by a study of in vitro co-culture CAR T cells with AN3CA (a human EC cell line) and in vivo AN3CA xenograft mice models. At the end of the treatment, when compared with 2 control groups, the experimental group which used MISIIR-specific CAR T cell therapy showed higher levels of IFN-γ in supernatants, approximately 2-fold shrinkage of tumor volume, higher concentrations of circulating CD3+T cells, and higher percentages of CD45+cells infiltrating in tumors.

Combination of adoptive T cell transfer with ICBs

Combining ACT with ICBs or traditional therapeutic methods may increase the potency of targeting and eliminating EC. In a phase I study (NCT03757858), Qiao et al. investigated several combination strategies of autologous ACT with other therapeutic methods in patients with advanced solid tumors. Thirty-three patients were divided into 3 therapeutic cohorts: 10 patients receiving hyperthermia plus ACT, 11 patients receiving hyperthermia plus ACT plus pembrolizumab and 12 patients receiving hyperthermia plus ACT plus chemotherapy. Hyperthermia can exert antitumor activity and have a synergistic effect with chemotherapy as a thermal sensitizer [289]. One EC patient was in the hyperthermia plus ACT cohort while both the hyperthermia plus ACT plus pembrolizumab and hyperthermia plus ACT plus chemotherapy cohort included 2 EC patients. Although the total ORR was 30% (10/33) and 3 patients achieved CR, the ORR for 5 EC patients was 20%, and no EC patient had a CR (1 PR, 2 SD and 2 PD for EC cohort). There was no immune related AEs in this trial with toxicities caused by chemotherapy and hyperthermia and mostly identified as G1-2(13/15 patients, 86.7%).

Immunotherapy in p53 mutant subtype

As mentioned earlier, the TCGA molecular classification [14] initially defined the molecular characteristics of the p53 mutant/serous-like subtype of EC and verified the genetic similarity of this type of EC with HGSOC, and basal-like breast carcinomas (BLBC) (account for 55-81% of triple-negative breast cancer, TNBC) [290, 291]. There are shared amplification mutations among the serous EC, HGSOC and BLBC such as MYC, ERBB3, CCNE1, MCL1, MECOM and FGF3 [14]. Additionally, serous EC patients have a higher frequency of ERBB2 amplification compared with those with HGSOC and BLBC, and most of the ERBB2 amplified EC cases have concurrent PIK3CA mutations [14]. Based on precision medicine, these molecular characteristics can guide clinical treatment including immunotherapy. It is increasingly appreciated that emerging treatment of HGSOC and TNBC can provide lessons for TP53 mutant EC, and separate consideration of novel strategies may benefit this EC subgroup [292].

For HGSOC [293] and TNBC [294], no official immunotherapy has been approved, and efforts are currently concentrated on the development of combination treatment of immunotherapy with other strategies. The phenotype of BRCAness is tightly associated with basal-like sporadic breast tumors and TNBC [295], and homologous recombination deficiency is observed in more than 50% of HGSOC patients [296]. Abnormal cell cycle related genes and the high frequency of TP53 mutations in copy-number high EC suggest vulnerability to DNA damage and repair dysfunction [292]. PARP inhibitors have shown great clinical activity in DNA repair deficient carcinomas. Recent experimental studies found that upregulation of PD-L1 induced by PARP inhibitors can produce immuno-tolerance during the cancer treatment, which can be blocked by PD1/PD-L1 antibodies. The TOPACIO (Keynote 162) trial (NCT02657889) is ongoing and is investigate the treatment of Niraparib (a type of PARP inhibitor) with pembrolizumab in patients with metastatic TNBC or OC. The data suggest adding PARP inhibitors to immunotherapy as a combination strategy may be potentially beneficial for copy-number high EC. Moreover, the mutation of PIK3CA gene, which encodes the PI3K catalytic subunit α and plays a role in PI3K/AKT/mTOR pathway, is frequently found in TNBC (10.2%) [295] and copy-number high EC [14]. The amplification of MYC, which can be targeted by MEK inhibitors, is also observed in a variety of serous EC, HGSOC [14], and 30% of TNBC or BLBC patients [297, 298]. A phase Ib trial (NCT02900664) investigating the treatment of advanced adenocarcinoma, including TNBC, with PD-L1 antibody plus MEK inhibitor is currently ongoing. Treatment-emergent AEs and dose limiting toxicities (DLTs) will be evaluated. These data suggest adding specific molecular targeted agents, such as PI3K/AKT/mTOR pathway and MEK inhibitors, to immunotherapy may provide powerful anti-tumor effects in copy-number high EC.

Recent studies have demonstrated that IDO is a critical molecular in inducing immuno-tolerance in HGSOC and TNBC. IDO1 positivity is found in 37% of all TNBCs and has a tight association with basal-like TNBC [299]. The African American Cancer Epidemiology Study (AACES) [300] demonstrated positive IDO expression in 58% of HGSOC, and most PD-L1-positive patients co-expressed IDO. Both of markers were associated with higher lymphocyte infiltration (P<0.05). Epacadostat (epac), an oral IDO inhibitor, can restore or promote the proliferation of dendritic cells, NK cells, and effector T cells as well as decreasing Treg cells. A combination treatment of epacadostat plus pembolizumab is being tested in a phase I/II, Keynote 037-ECHO 202 study (NCT 02178722) in patients with selected cancers including TNBC. Although there are limited data about the expression status and related treatment of IDO in copy-number high EC, it is important to explore the IDO related immunotherapy in this subgroup.

Amplification of ERBB2 (17q12) is found in 26-62% of uterine serous cancer patients [301,302,303,304] and has been defined as one of the significant amplified oncogenes in copy-number high EC [14]. Thus, human epidermal growth factor receptor 2 (HER2/Neu, also known as ERBB2) targeted immunotherapy has been considered as a powerful treatment for this subtype of EC. This kind of treatment employs humanized monoclonal antibodies targeting HER2, such as trastuzumab and pertuzumab, to recruit NK cells via Fc region conjugation [305] and kill the tumor cells through antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) effects [306,307,308]. However, in a GOG-181B trial [309] investigating the efficacy of trastuzumab monotherapy in patients with stage III/IV, recurrent, HER2-positive EC, trastuzumab showed no clinical activity in EC with HER2 overexpression or amplification. However, only 28% (7/25) of serous carcinoma patients were HER2 positive and able to be enrolled in this trial. Recently, Fader et al. [310] published the results analysis of a multicenter, randomized phase II trial of treating HER2 overexpressed advanced serous EC with carboplatin-paclitaxel (control) or carboplatin-paclitaxel plus trastuzumab (experimental). Fifty-eight patients were evaluated, and the median PFS in the control group vs experimental group was 8.0 vs 12.6 months (P=0.005; hazard ratio [HR], 0.44; 90% CI, 0.26 to 0.76), suggesting improved outcomes with the addition of trastuzumab to basic chemotherapy. Toxicity was not different between the control and experimental groups.

BiTE antibody (bispecific antibody) is a novel drug which bridges cancer cells with cytotoxic T cells and induces a direct cytolytic effect of T cells without the restriction of specific T cell receptor, MHC class I molecules, or peptide antigen presentation [311,312,313]. Bellone et al [314] performed a preclinical study using BiTE antibodies (solitomab) in patients with epithelial-cell-adhesion-molecule (EpCAM) over-expressed uterine serous EC(USC). EpCAM expression is found in 87.5% of USC which has been demonstrated to be susceptible to solitomab, a type of EpCAM/CD3 bispecific antibody. The clinical efficacy of solitomab has been noted in multiple cancers such as colon cancer and OC [315, 316]. In this preclinical study of EC, the solitomab treated group displayed increased cytokine secretion, T lymphocyte activation and proliferation, as well as cytotoxic activity of tumor associated lymphocytes, suggesting clinical value of this drug [314].

MetS & immunotherapy

MetS represents a group of risk factors related to the development of diabetes and cardiovascular disease [317]. Obesity, diabetes and hypertension, as the common phenotype of MetS, are often found as a metabolic triad in EC patients [318]. The risk of EC in overweight (BMI≥ 25 kg/m2), diabetic, or hypertensive patients were, respectively, 2.45, 2.12, and 3.5 times higher than control groups [318]. Based on a series of retrospective and prospective studies, EC is now defined as one of the tumor types most closely associated with MetS [1]. MetS can promote EC development through complex modulating mechanisms, local inflammation [319], and remodeled immune microenvironment [318, 320].

Obesity is one of the key characteristics of MetS and adipose tissue plays an essential role in the pathophysiological changes and tumor-microenvironment interaction in EC [320]. In obese people, the balance between adipocytes and immune cells is impaired. Obesity-related chronic adipose inflammation gradually develops [321] and promotes tumor progression [322, 323]. A cluster of pro-inflammatory cytokines, such as IL-6, TNF-α and IL-18, are secreted by adipocytes to facilitate the infiltration of lymphocytes in EC cells [324], resulting in angiogenesis [325], tissue remodeling [319], and a pro-neoplastic microenvironment [321]. The local inflammatory mechanism promotes malignant transformation of normal tissues and tumor development [326].

Systemic hyperglycemia is a characteristic of diabetes which serves as a favorable metabolic environment for the rapid proliferation of cancer cells [318]. In obese EC patients, cancer cells produce high levels of lactic acid (a metabolite of glycolysis) during hyperglycemia, which can promote malignant transformation of normal cells through metabolism re-programming [318] and also transform anti-tumor M1 macrophages to pro-tumor M2 macrophages [327]. Insulin resistance coupled with hyperinsulinemia and up-regulated insulin growth factor-1 are not only seen in diabetes but also has a tight association with hyperglycemia, obesity, and cancer development [328].In MetS patients, obesity-related inflammation induces the expression of a cluster of inflammatory cytokines including C-reactive protein (CRP), IL-6, TGF-α, and plasminogen activator inhibitor-1 (PAI-1) [329, 330], which further influence insulin signaling pathways and lead to insulin resistance. Elevated insulin and IGF-1 can combine with IR and IGF-1 receptors (IGF-1R) respectively and promote EC cell proliferation via downstream signal transformation [331]. Metformin is a type of insulin-sensitizing anti-hyperglycemic drug which is widely used in treating type II diabetes mellitus [332, 333]. Several new applications for this agent have recently been found, one of which is its anti-tumor effects [334]. Metformin has shown promising efficacy as a new adjunctive treatment in EC. A systematic review and meta-analysis raised by Meireles et al. [335] demonstrated that metformin treatment led to reversion of atypical endometrial hyperplasia to normal endometrium with down-regulated proliferation markers (from 51.94% to 34.47%, CI = 36.23-67.46% and 18.55-52.43% ). Higher OS was also observed in metformin-treated EC patients compared with non-metformin-treated and non-diabetic patients (HR =0.82; CI: 0.70-0.95; P=0.09, I [2]=40% ). Prior studies have identified several anti-tumor mechanisms of metformin [336,337,338] with adenosine monophosphate-activated protein kinase (AMPK)-dependent PI3K/Akt/mTOR pathway inhibition considered as an important direct mechanism of metformin in treating EC [333, 339]. Furthermore, metformin also has immune-mediated antitumor effect including blocking the PD-L1/PD-1 axis [340], increasing CD8+TIL infiltration, and protecting CD8+TILs from apoptosis and exhaustion [341]. Based on its immune-related effects, preclinical studies have found synergistic antitumor effects by combining metformin with cancer vaccines [341] and CTLA4 blockades [340]. Though there is no clinical study testing metformin combination immunotherapy, it is promising to explore the efficacy of this combination strategy.

Hormone & immunotherapy

Hormone aberrations, such as estrogenic excess, lack of progesterone and abnormal expression of endometrial receptors, have long been considered as significant etiological factors for EC [342, 343]. Elevated estrogen results in angiogenesis, endometrial cell proliferation and apoptosis inhibition [344], thus promoting carcinogenesis. Progesterone deficiency decrease its protective effects on endometrial epithelial cells from malignant transformation, such as inducing cells apoptosis via binding to PR [345], inhibiting ERα expression, and controlling growth factor production of stromal cells [346, 347]. Sex hormones also have immuno-modulatory functions which participate in tumor-immunity interactions. Almost all types of immune cells express receptors for progesterone and estrogen [348,349,350,351], and ER and androgen (AR) responsive elements have been found located on the promoters of many immune-related genes [352]. In fact, one sex hormone can have both immune stimulatory and immune inhibitory functions based on the doses and time of action [353]. Estrogen can both promote inflammation through inducing IFN-γ and IL-2 secretion by peripheral T cells [354] and also induce immune tolerance via stimulating IL-10 production [355]. Progesterone can enhance humoral immune responses and the production of IL-5, IL-6, and IL-10 [356], while also inhibiting T cell proliferation [357] and IFN-γ production [358]. Witkiewicz et al [359] investigated the use of progestin in 15 patients with complex atypical endometrial hyperplasia and well-differentiated EC who desired fertility preservation or were unsuitable for surgery. After completion of the treatment, 66.7% (10/15) of patients had normal morphology on follow-up sampling while 26.7% (4/15) of patients had persistent or progressive disease. Progestin was found to significantly influence the subpopulations of lymphocytes as decreased Tregs and increased NK cells were observed in post-treatment specimens. Because of this complex immune-regulatory mechanism about which research has been limited the roles of sex hormones in immuno-oncology still remain to be identified in EC.

In EC patients, deficiency of both ER and PR expression is an independent prognostic marker for worse outcomes, even in those defined as low-grade [360]. The double negative of ER and PR is also considered a predictor for lymph node metastases [361] and tumor relapse [362]. Furthermore, single deficiency of PR expression predicts poor outcome in high-grade EC patients, even in those whose histotypes, such as serous type, were previously recognized as hormone-independent types [363]. The ER-positive rates are similar among the four TCGA molecular subtypes (POLE:75.7%, MSI:73.9%, TP5 WT:92%, TP53 mutant:67.4%), while the PR-positive rate in TP53 WT group is the highest (POLE: 75%, MSI: 60.9, TP53 WT: 83.9%, TP53 mutant: 44.7%) [364]. An analysis of early stage EC in the PORTEC cohorts showed a higher ER/PR-negative rate in TP53 mutant group [365]. These data provide basis for stratifying clinical risks within different molecular phenotypes. Recent studies have proposed controversial opinions on the relationship between ER and PR expression status and immune responses in EC. Jiang et al. [366] found that the infiltration of tumor-associated macrophages was higher in PR-negative EC cases compared with positive ones (P = 0.0001), which predicted immuno-tolerance of tumors and poor outcomes. However, Giatromanolaki et al. [367] suggested that a low infiltration of FOXP3+ Treg cells, which participate in pro-tumor immune responses, was associated with ER-negative and low vascular density in EC.

Ongoing trials of endocrine therapy are fewer than those of chemotherapy and new target therapies [368]; however, endocrine therapy still warrants attention due to its good tolerance and known toxicity [368]. The combination of endocrine therapy with immunotherapy may be an alternative option for young patients seeking fertility sparing therapies. A phase 1 trial (NCT04046185) is now ongoing to investigate the combination of Toripalimab with progesterone in Stage I, FIGO grade 1-2 endometrioid EC patients who desire fertility preservation. The pathologic complete/partial remission rates will be evaluated at 6 months after the treatment initiation.

Conclusion

In 1893, William Coley, a New York orthopedic surgeon, accidentally observed that the tumor tissue of patients infected with Streptococcus pyogenes could make the tumor tissue of some patients slowly recede, which opened the prelude of tumor immunotherapy [369]. Anti-tumor immunotherapy has achieved ideal results in the treatment of malignant tumors, but there are still many problems in clinical application that have not been resolved. The reason is that the understanding of tumor immune mechanism is not thorough enough currently, and more research is needed to reveal new cellular and molecular mechanisms. For the problems that arise in immunotherapy, it is first necessary to formulate individualized treatment strategies to achieve individualized precision treatment. Secondly, it is necessary to establish a set of reasonable evaluation standards reflecting the effect of immunotherapy. Finally, it is necessary to overcome tumor immune tolerance. While using the activated immune system to treat tumors, it is necessary to reduce the immunosuppressive environment in patients, especially the immunosuppressive microenvironment inside tumor tissues. With the accumulation of more clinical experience and the development of scientific research, we will have a deeper understanding of tumor immunotherapy, and we have reason to believe that this novel treatment method can be applied to cancer patients more safely and effectively.

Availability of data and materials

Not applicable.

Abbreviations

ICIs:

Immune checkpoint inhibitors

ACT:

Adoptive cell transfer

EC:

Endometrial cancer

MetS:

Metabolic syndrome

ICBs:

Immune checkpoint blockades

RT:

Radiotherapy

POLE:

Polymerase-ε

MSI-H:

Microsatellite instability hypermutated

MMR:

Mismatch repair

MSS:

Microsatellite stability

HGSOC:

High-grade serous ovarian carcinoma

NK:

Natural killer

Treg:

Regulatory T cells

DCs:

Dendritic cells

CTLs:

CD8+ cytotoxic T cells

IDO:

Indoleamine 2, 3-dioxygenase

MDSC:

Myeloid derived suppressor cells

APC:

Antigen-presenting cell

PD1:

Programmed cell death 1

CTLA4:

Cytotoxic T lymphocyte antigen 4

PDL1:

Programmed cell death 1 ligand 1

NSCLC:

Non-small-cell lung cancer

FDA:

Food and drug administration

OS:

Overall survival

SCLC:

Small-cell lung cancer

ORR:

Objective response rate

TILs:

Tumor-infiltrating lymphocytes

OC:

Ovarian cancer

DFS:

Disease free survival

LS:

Lynch syndrome

COX-2:

Cyclooxygenase-2

IFN-γ:

Interferon-γ

ARID1:

AT-rich interaction domain 1A

CR:

Complete response

SD:

Stable disease

AEs:

Adverse events

PD:

Progressive disease

rEC:

Recurrent endometrial cancer

IC:

immune cells

DOR:

Duration of objective response

OR:

Objective response

PFS6:

PFS at 6 months

OTR:

Objective tumor response

AEC:

Advanced endometrial cancer

PARP:

Poly (ADP-ribose) polymerase

ADCs:

Antibody-drug conjugate

LAG-3:

Lymphocyte-activation gene 3

MTD:

Maximum tolerated dose

RD:

Recommended dose

TIM-3:

T cell immunoglobulin and mucin-domain containing-3

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

HER2:

Human epidermal growth factor receptor 2

FSPs:

Frameshift peptides

LAK:

Lymphokine-activated killer

IL-2:

Interleukin-2

rIL-2:

Recombinant IL-2

CAR:

Chimeric antigen receptors

MISIIR:

Müllerian inhibiting substance type II receptor

BLBC:

Basal-like breast carcinomas

TNBC:

Triple-negative breast cancer

DLTs:

Dose limiting toxicities

EpCAM:

Epithelial-cell-adhesion-molecule

BC:

Breast cancer

CC:

Cervical cancer

References

  1. Morice P, Leary A, Creutzberg C, Abu-Rustum N, Darai E. Endometrial cancer. Lancet. 2016;387(10023):1094–108.

    Article  PubMed  Google Scholar 

  2. Siegel R, Miller K, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7–30.

    Article  PubMed  Google Scholar 

  3. Lortet-Tieulent J, Ferlay J, Bray F, Jemal A. International Patterns and Trends in Endometrial Cancer Incidence, 1978-2013. J Natl Cancer Inst. 2018;110(4):354–61.

    Article  PubMed  Google Scholar 

  4. Clarke M, Devesa S, Harvey S, Wentzensen N. Hysterectomy-Corrected Uterine Corpus Cancer Incidence Trends and Differences in Relative Survival Reveal Racial Disparities and Rising Rates of Nonendometrioid Cancers. J Clin Oncol. 2019;37(22):1895–908.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Connor E, Rose P. Management Strategies for Recurrent Endometrial Cancer. Expert Rev Anticancer Ther. 2018;18(9):873–85.

    Article  CAS  PubMed  Google Scholar 

  6. Wolchok J, Chiarion-Sileni V, Gonzalez R, et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med. 2017;377(14):1345–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Motzer R, Tannir N, McDermott D, et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med. 2018;378(14):1277–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Maude S, Laetsch T, Buechner J, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018;378(5):439–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Di Tucci C, Capone C, Galati G, et al. Immunotherapy in endometrial cancer: new scenarios on the horizon. J Gynecol Oncol. 2019;30(3):e46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nishio H, Iwata T, Aoki D. Current status of cancer immunotherapy for gynecologic malignancies. Jpn J Clin Oncol. 2021;51(2):167–72.

    Article  PubMed  Google Scholar 

  11. Di Tucci C, Schiavi MC, Faiano P, et al. Therapeutic vaccines and immune checkpoints inhibition options for gynecological cancers. Crit Rev Oncol Hematol. 2018;128:30–42.

    Article  PubMed  Google Scholar 

  12. Yang Y. Cancer immunotherapy: harnessing the immune system to battle cancer. J Clin Invest. 2015;125(9):3335–7.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Riley R, June C, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov. 2019;18(3):175–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kandoth C, Schultz N, Cherniack A, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67–73.

    Article  PubMed  CAS  Google Scholar 

  15. Briggs S, Tomlinson I. Germline and somatic polymerase ε and δ mutations define a new class of hypermutated colorectal and endometrial cancers. J Pathol. 2013;230(2):148–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Church D, Stelloo E, Nout R, et al. Prognostic significance of POLE proofreading mutations in endometrial cancer. J Natl Cancer Inst. 2015;107(1):402.

    Article  PubMed  CAS  Google Scholar 

  17. Meng B, Hoang L, McIntyre J, et al. POLE exonuclease domain mutation predicts long progression-free survival in grade 3 endometrioid carcinoma of the endometrium. Gynecol Oncol. 2014;134(1):15–9.

    Article  CAS  PubMed  Google Scholar 

  18. Zhao S, Choi M, Overton J, et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A. 2013;110(8):2916–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Stelloo E, Bosse T, Nout R, et al. Refining prognosis and identifying targetable pathways for high-risk endometrial cancer; a TransPORTEC initiative. Mod Pathol. 2015;28(6):836–44.

    Article  CAS  PubMed  Google Scholar 

  20. McConechy M, Talhouk A, Leung S, et al. Endometrial Carcinomas with POLE Exonuclease Domain Mutations Have a Favorable Prognosis. Clin Cancer Res. 2016;22(12):2865–73.

    Article  CAS  PubMed  Google Scholar 

  21. Bellone S, Bignotti E, Lonardi S, et al. Polymerase ε (POLE) ultra-mutation in uterine tumors correlates with T lymphocyte infiltration and increased resistance to platinum-based chemotherapy in vitro. Gynecol Oncol. 2017;144(1):146–52.

    Article  CAS  PubMed  Google Scholar 

  22. Rayner E, van Gool I, Palles C, et al. A panoply of errors: polymerase proofreading domain mutations in cancer. Nat Rev Cancer. 2016;16(2):71–81.

    Article  CAS  PubMed  Google Scholar 

  23. Le Gallo M, Bell D. The emerging genomic landscape of endometrial cancer. Clin Chem. 2014;60(1):98–110.

    Article  PubMed  CAS  Google Scholar 

  24. Murali R, Soslow R, Weigelt B. Classification of endometrial carcinoma: more than two types. Lancet Oncol. 2014;15(7):e268–78.

    Article  PubMed  Google Scholar 

  25. Richman S. Deficient mismatch repair: Read all about it (Review). Int J Oncol. 2015;47(4):1189–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Diaz-Padilla I, Romero N, Amir E, et al. Mismatch repair status and clinical outcome in endometrial cancer: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2013;88(1):154–67.

    Article  PubMed  Google Scholar 

  27. Kanopienė D, Smailytė G, Vidugirienė J, Bacher J. Impact of microsatellite instability on survival of endometrial cancer patients. Medicina (Kaunas). 2014;50(4):216–21.

    Article  PubMed  Google Scholar 

  28. Howitt B, Shukla S, Sholl L, et al. Association of Polymerase e-Mutated and Microsatellite-Instable Endometrial Cancers With Neoantigen Load, Number of Tumor-Infiltrating Lymphocytes, and Expression of PD-1 and PD-L1. JAMA Oncol. 2015;1(9):1319–23.

    Article  PubMed  Google Scholar 

  29. Eggink F, Van Gool I, Leary A, et al. POLEImmunological profiling of molecularly classified high-risk endometrial cancers identifies -mutant and microsatellite unstable carcinomas as candidates for checkpoint inhibition. Oncoimmunology. 2017;6(2):e1264565.

    Article  PubMed  CAS  Google Scholar 

  30. Gargiulo P, Della Pepa C, Berardi S, et al. Tumor genotype and immune microenvironment in POLE-ultramutated and MSI-hypermutated Endometrial Cancers: New candidates for checkpoint blockade immunotherapy? Cancer Treat Rev. 2016;48:61–8.

    Article  CAS  PubMed  Google Scholar 

  31. Raffone A, Travaglino A, Mascolo M, et al. TCGA molecular groups of endometrial cancer: Pooled data about prognosis. Gynecol Oncol. 2019;155(2):374–83.

    Article  CAS  PubMed  Google Scholar 

  32. Cosgrove C, Tritchler D, Cohn D, et al. An NRG Oncology/GOG study of molecular classification for risk prediction in endometrioid endometrial cancer. Gynecol Oncol. 2018;148(1):174–80.

    Article  CAS  PubMed  Google Scholar 

  33. Vanderstraeten A, Tuyaerts S, Amant F. The immune system in the normal endometrium and implications for endometrial cancer development. J Reprod Immunol. 2015;109:7–16.

    Article  CAS  PubMed  Google Scholar 

  34. Wira C, Grant-Tschudy K, Crane-Godreau M. Epithelial cells in the female reproductive tract: a central role as sentinels of immune protection. Am J Reprod Immunol. 2005;53(2):65–76.

    Article  CAS  PubMed  Google Scholar 

  35. Ochiel D, Ghosh M, Fahey J, Guyre P, Wira C. Human uterine epithelial cell secretions regulate dendritic cell differentiation and responses to TLR ligands. J Leukoc Biol. 2010;88(3):435–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ferguson A, Moore M. Fox HJBjoc. Expression of MHC products and leucocyte differentiation antigens in gynaecological neoplasms: an immunohistological analysis of the tumour cells and infiltrating leucocytes. Br J Cancer. 1985;52(4):551–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wira C, Fahey J, Ghosh M, Patel M, Hickey D, Ochiel D. Sex hormone regulation of innate immunity in the female reproductive tract: the role of epithelial cells in balancing reproductive potential with protection against sexually transmitted pathogens. Am J Reprod Immunol. 2010;63(6):544–65.

    Article  CAS  PubMed  Google Scholar 

  38. Longoria T, Eskander R. Immunotherapy in endometrial cancer - an evolving therapeutic paradigm. Gynecol Oncol Res Pract. 2015;2:11.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hickey D, Patel M, Fahey J, Wira C. Innate and adaptive immunity at mucosal surfaces of the female reproductive tract: stratification and integration of immune protection against the transmission of sexually transmitted infections. J Reprod Immunol. 2011;88(2):185–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Manaster I, Mizrahi S, Goldman-Wohl D, et al. Endometrial NK cells are special immature cells that await pregnancy. J Immunol. 2008;181(3):1869–76.

    Article  CAS  PubMed  Google Scholar 

  41. Yeaman G, Guyre P, Fanger M, et al. Unique CD8+ T cell-rich lymphoid aggregates in human uterine endometrium. J Leukoc Biol. 1997;61(4):427–35.

    Article  CAS  PubMed  Google Scholar 

  42. Yeaman G, Collins J, Fanger M, Wira C, Lydyard P. CD8+ T cells in human uterine endometrial lymphoid aggregates: evidence for accumulation of cells by trafficking. Immunology. 2001;102(4):434–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Marshall R, Jones D. An immunohistochemical study of lymphoid tissue in human endometrium. Int J Gynecol Pathol. 1988;7(3):225–35.

    Article  CAS  PubMed  Google Scholar 

  44. Wira C, Fahey J, Sentman C, Pioli P, Shen L. Innate and adaptive immunity in female genital tract: cellular responses and interactions. Immunol Rev. 2005;206:306–35.

    Article  PubMed  Google Scholar 

  45. Dunn G, Bruce A, Ikeda H, Old L, Schreiber R. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3(11):991–8.

    Article  CAS  PubMed  Google Scholar 

  46. Burnet M. Cancer: a biological approach. III. Viruses associated with neoplastic conditions. IV. Practical applications. Br Med J. 1957;1(5023):841–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Stutman O. Immunodepression and malignancy. Adv Cancer Res. 1975;22:261–422.

    Article  CAS  PubMed  Google Scholar 

  48. Stutman O. Tumor development after 3-methylcholanthrene in immunologically deficient athymic-nude mice. Science. 1974;183(4124):534–6.

    Article  CAS  PubMed  Google Scholar 

  49. Ikeda H, Old L, Schreiber RJC. reviews gf. The roles of IFN gamma in protection against tumor development and cancer immunoediting. Cytokine Growth F R. 2002;13(2):95–109.

    Article  CAS  Google Scholar 

  50. O'Donnell J, Teng M, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol. 2019;16(3):151–67.

    Article  CAS  PubMed  Google Scholar 

  51. Grivennikov S, Greten F, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mellman I, Steinman RM. Dendritic cells: specialized and regulated antigen processing machines. Cell. 2001;106(3):255–8.

    Article  CAS  PubMed  Google Scholar 

  53. Palucka K, Banchereau J, Mellman I. Designing vaccines based on biology of human dendritic cell subsets. Immunity. 2010;33(4):464–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chimal-Ramírez G, Espinoza-Sánchez N, Fuentes-Pananá EM. Protumor activities of the immune response: insights in the mechanisms of immunological shift, oncotraining, and oncopromotion. J Oncol. 2013;2013:835956.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Schreiber R, Old L, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science. 2011;331(6024):1565–70.

    Article  CAS  PubMed  Google Scholar 

  56. Quezada S, Peggs K, Simpson T, Allison JP. Shifting the equilibrium in cancer immunoediting: from tumor tolerance to eradication. Immunol Rev. 2011;241(1):104–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Aguirre-Ghiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer. 2007;7(11):834–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Koebel C, Vermi W, Swann J, et al. Adaptive immunity maintains occult cancer in an equilibrium state. Nature. 2007;450(7171):903–7.

    Article  CAS  PubMed  Google Scholar 

  59. Gabrilovich D, Ishida T, Oyama T, et al. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998;92(11):4150–66.

    Article  CAS  PubMed  Google Scholar 

  60. Yoshimura A, Muto G. TGF-β function in immune suppression. Curr Top Microbiol Immunol. 2011;350:127–47.

    CAS  PubMed  Google Scholar 

  61. Löb S, Königsrainer A, Rammensee H, Opelz G, Terness P. Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: can we see the wood for the trees? Nat Rev Cancer. 2009;9(6):445–52.

    Article  PubMed  CAS  Google Scholar 

  62. Garrido F, Ruiz-Cabello F, Cabrera T, et al. Implications for immunosurveillance of altered HLA class I phenotypes in human tumours. IMMUNOL TODAY. 1997;18(2):89–95.

    Article  CAS  PubMed  Google Scholar 

  63. Ventriglia J, Paciolla I, Pisano C, et al. Immunotherapy in ovarian, endometrial and cervical cancer: State of the art and future perspectives. Cancer Treat Rev. 2017;59:109–16.

    Article  CAS  PubMed  Google Scholar 

  64. Bretscher PA. A two-step, two-signal model for the primary activation of precursor helper T cells. Proc Natl Acad Sci U S A. 1999;96(1):185–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Khalil D, Smith E, Brentjens R, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016;13(5):273–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Appleman L, Boussiotis VA. T cell anergy and costimulation. Immunol Rev. 2003;192:161–80.

    Article  CAS  PubMed  Google Scholar 

  67. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348(6230):56–61.

    Article  CAS  PubMed  Google Scholar 

  68. Townsend S, Allison JP. Tumor rejection after direct costimulation of CD8+ T cells by B7-transfected melanoma cells. Science. 1993;259(5093):368–70.

    Article  CAS  PubMed  Google Scholar 

  69. Wei S, Duffy C, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018;8(9):1069–86.

    Article  PubMed  Google Scholar 

  70. Linsley P, Greene J, Brady W, Bajorath J, Ledbetter J, Peach R. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity. 1994;1(9):793–801.

    Article  CAS  PubMed  Google Scholar 

  71. Linsley P, Brady W, Urnes M, Grosmaire L, Damle N, Ledbetter JA. CTLA-4 is a second receptor for the B cell activation antigen B7. J Exp Med. 1991;174(3):561–9.

    Article  CAS  PubMed  Google Scholar 

  72. Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209(6):1201–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Paterson A, Brown K, Keir M, et al. The programmed death-1 ligand 1:B7-1 pathway restrains diabetogenic effector T cells in vivo. J Immunol. 2011;187(3):1097–105.

    Article  CAS  PubMed  Google Scholar 

  74. Blank C, Gajewski T, Mackensen A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: implications for tumor immunotherapy. Cancer Immunol Immunother. 2005;54(4):307–14.

    Article  CAS  PubMed  Google Scholar 

  75. Patel S, Kurzrock R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol Cancer Ther. 2015;14(4):847–56.

    Article  CAS  PubMed  Google Scholar 

  76. Pakish J, Zhang Q, Chen Z, et al. Immune Microenvironment in Microsatellite-Instable Endometrial Cancers: Hereditary or Sporadic Origin Matters. Clin Cancer Res. 2017;23(15):4473–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Crumley S, Kurnit K, Hudgens C, et al. Identification of a subset of microsatellite-stable endometrial carcinoma with high PD-L1 and CD8+ lymphocytes. Mod Pathol. 2019;32(3):396–404.

    Article  CAS  PubMed  Google Scholar 

  78. Luchini C, Bibeau F, Ligtenberg M, et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol. 2019;30(8):1232–43.

    Article  CAS  PubMed  Google Scholar 

  79. Vanderwalde A, Spetzler D, Xiao N, Gatalica Z, Marshall J. Microsatellite instability status determined by next-generation sequencing and compared with PD-L1 and tumor mutational burden in 11,348 patients. Cancer Med. 2018;7(3):746–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Reck M, Rodríguez-Abreu D, Robinson A, et al. Pembrolizumab versus Chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375(19):1823–33.

    Article  CAS  PubMed  Google Scholar 

  81. Herbst R, Baas P, Kim D, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540–50.

    Article  CAS  PubMed  Google Scholar 

  82. Bellmunt J, de Wit R, Vaughn D, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med. 2017;376(11):1015–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Muro K, Chung H, Shankaran V, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol. 2016;17(6):717–26.

    Article  CAS  PubMed  Google Scholar 

  84. Fuchs C, Doi T, Jang R, et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients With Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol. 2018;4(5):e180013.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Powles T, Durán I, van der Heijden M, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748–57.

    Article  CAS  PubMed  Google Scholar 

  86. Ferris R, Blumenschein G, Fayette J, et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N Engl J Med. 2016;375(19):1856–67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Ott P, Elez E, Hiret S, et al. Pembrolizumab in Patients With Extensive-Stage Small-Cell Lung Cancer: Results From the Phase Ib KEYNOTE-028 Study. J Clin Oncol. 2017;35(34):3823–9.

    Article  CAS  PubMed  Google Scholar 

  88. McLaughlin J, Han G, Schalper K, et al. Quantitative Assessment of the Heterogeneity of PD-L1 Expression in Non-Small-Cell Lung Cancer. JAMA Oncol. 2016;2(1):46–54.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Goyal G, Lau D, Nagle A, et al. Tumor mutational burden and other predictive immunotherapy markers in histiocytic neoplasms. Blood. 2019;133(14):1607–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Samstein R, Lee C, Shoushtari A, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51(2):202–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Chalmers Z, Connelly C, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Rizvi H, Sanchez-Vega F, La K, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. J Clin Oncol. 2018;36(7):633–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Tubbs A, Nussenzweig A. Endogenous DNA Damage as a Source of Genomic Instability in Cancer. Cell. 2017;168(4):644–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Perumal D, Imai N, Laganà A, et al. Mutation-derived Neoantigen-specific T-cell Responses in Multiple Myeloma. Clin Cancer Res. 2020;26(2):450–64.

    Article  CAS  PubMed  Google Scholar 

  95. Peggs K, Segal N, Allison JP. Targeting immunosupportive cancer therapies: accentuate the positive, eliminate the negative. Cancer Cell. 2007;12(3):192–9.

    Article  CAS  PubMed  Google Scholar 

  96. Segal N, Parsons D, Peggs K, et al. Epitope landscape in breast and colorectal cancer. Cancer Res. 2008;68(3):889–92.

    Article  CAS  PubMed  Google Scholar 

  97. Chan T, Wolchok J, Snyder A. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2015;373(20):1984.

    Article  CAS  PubMed  Google Scholar 

  98. Matsushita H, Vesely M, Koboldt D, et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature. 2012;482(7385):400–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Riaz N, Morris L, Havel J, Makarov V, Desrichard A, Chan T. The role of neoantigens in response to immune checkpoint blockade. Int Immunol. 2016;28(8):411–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Ott P, Hu Z, Keskin D, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547(7662):217–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Cohen C, Gartner J, Horovitz-Fried M, et al. Isolation of neoantigen-specific T cells from tumor and peripheral lymphocytes. J Clin Invest. 2015;125(10):3981–91.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371(23):2189–99.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Hellmann M, Nathanson T, Rizvi H, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 2018;33(5):843–852.e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Le D, Durham J, Smith K, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Le D, Uram J, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Lauss M, Donia M, Harbst K, et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat Commun. 2017;8(1):1738.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Zaretsky J, Garcia-Diaz A, Shin D, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med. 2016;375(9):819–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Skoulidis F, Goldberg M, Greenawalt D, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in -mutant lung adenocarcinoma. Cancer Discov. 2018;8(7):822–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Carbone D, Reck M, Paz-Ares L, et al. First-line nivolumab in stage iv or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376(25):2415–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Hellmann M, Ciuleanu T, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378(22):2093–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Goodman A, Kato S, Bazhenova L, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16(11):2598–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Peters S, Gettinger S, Johnson M, et al. Phase II trial of atezolizumab as first-line or subsequent therapy for patients with programmed death-ligand 1-selected advanced non-small-cell lung cancer (BIRCH). [published correction appears in J Clin Oncol. 2018 Mar 20;36(9):931]. J Clin Oncol.2017;35(24):2781–9.

  113. Clark W, From L, Bernardino E, Mihm M. The histogenesis and biologic behavior of primary human malignant melanomas of the skin. J Clin Oncol. 1969;29(3):705–27.

    Google Scholar 

  114. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44.

    Article  CAS  PubMed  Google Scholar 

  115. Lee N, Zakka L, Mihm M, Schatton T. Tumour-infiltrating lymphocytes in melanoma prognosis and cancer immunotherapy. Pathology. 2016;48(2):177–87.

    Article  PubMed  Google Scholar 

  116. Sudo T, Nishida R, Kawahara A, et al. clinical impact of tumor-infiltrating lymphocytes in esophageal squamous cell carcinoma. Ann Surg Oncol. 2017;24(12):3763–70.

    Article  PubMed  Google Scholar 

  117. Stanton S, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J Immunother Cancer. 2016;4:59.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Duong C, Yong C, Kershaw M, Slaney C, Darcy PK. Cancer immunotherapy utilizing gene-modified T cells: From the bench to the clinic. Mol Immunol. 2015;67:46–57.

    Article  CAS  PubMed  Google Scholar 

  119. Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348(3):203–13.

    Article  CAS  PubMed  Google Scholar 

  120. Sato E, Olson S, Ahn J, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102(51):18538–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. de Jong R, Leffers N, Boezen H, et al. Presence of tumor-infiltrating lymphocytes is an independent prognostic factor in type I and II endometrial cancer. Gynecol Oncol. 2009;114(1):105–10.

    Article  PubMed  CAS  Google Scholar 

  122. Tumeh P, Harview C, Yearley J, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the 'Immunoscore' in the classification of malignant tumours. J Pathol. 2014;232(2):199–209.

    Article  CAS  PubMed  Google Scholar 

  124. Angell H, Galon J. From the immune contexture to the Immunoscore: the role of prognostic and predictive immune markers in cancer. Curr Opin Immunol. 2013;25(2):261–7.

    Article  CAS  PubMed  Google Scholar 

  125. Galon J, Pagès F, Marincola F, et al. The immune score as a new possible approach for the classification of cancer. J Transl Med. 2012;10:1.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Woerner S, Gebert J, Yuan Y, et al. Systematic identification of genes with coding microsatellites mutated in DNA mismatch repair-deficient cancer cells. Int J Cancer. 2001;93(1):12–9.

    Article  CAS  PubMed  Google Scholar 

  127. Maby P, Tougeron D, Hamieh M, et al. Correlation between Density of CD8+ T-cell Infiltrate in Microsatellite Unstable Colorectal Cancers and Frameshift Mutations: A Rationale for Personalized Immunotherapy. Cancer Res. 2015;75(17):3446–55.

    Article  CAS  PubMed  Google Scholar 

  128. Auguste A, Genestie C, De Bruyn M, et al. Refinement of high-risk endometrial cancer classification using DNA damage response biomarkers: a TransPORTEC initiative. Mod Pathol. 2018;31(12):1851–61.

    Article  CAS  PubMed  Google Scholar 

  129. Llosa N, Cruise M, Tam A, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5(1):43–51.

    Article  CAS  PubMed  Google Scholar 

  130. Phillips S, Banerjea A, Feakins R, Li S, Bustin S, Dorudi S. Tumour-infiltrating lymphocytes in colorectal cancer with microsatellite instability are activated and cytotoxic. Br J Surg. 2004;91(4):469–75.

    Article  CAS  PubMed  Google Scholar 

  131. Liu M, Wang X, Wang L, et al. Targeting the IDO1 pathway in cancer: from bench to bedside. J Hematol Oncol. 2018;11(1):100.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Cheong J, Ekkati A, Sun LJ. A patent review of IDO1 inhibitors for cancer. Expert Opin Ther Pat. 2018;28(4):317–30.

    Article  CAS  PubMed  Google Scholar 

  133. de Jong R, Kema I, Boerma A, et al. Prognostic role of indoleamine 2,3-dioxygenase in endometrial carcinoma. Gynecol Oncol. 2012;126(3):474–80.

    Article  PubMed  CAS  Google Scholar 

  134. Vanderstraeten A, Luyten C, Verbist G, Tuyaerts S, Amant F. Mapping the immunosuppressive environment in uterine tumors: implications for immunotherapy. Cancer Immunol Immunother. 2014;63(6):545–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Mills A, Zadeh S, Sloan E, et al. Indoleamine 2,3-dioxygenase in endometrial cancer: a targetable mechanism of immune resistance in mismatch repair-deficient and intact endometrial carcinomas. Mod Pathol. 2018;31(8):1282–90.

    Article  CAS  PubMed  Google Scholar 

  136. Lambropoulou M, Alexiadis G, Limberis V, Nikolettos N, Tripsianis G. Clinicopathologic and prognostic significance of cyclooxygenase-2 expression in endometrial carcinoma. Histol Histopathol. 2005;20(3):753–9.

    CAS  PubMed  Google Scholar 

  137. Cai SN, Zhang YX, Han K, Ding YQ. Expressions and clinical significance of COX-2, VEGF-C, and EFGR in endometrial carcinoma. Arch Gynecol Obstet. 2017;296(1):93–8.

    Article  CAS  PubMed  Google Scholar 

  138. Fosslien E. Molecular pathology of cyclooxygenase-2 in neoplasia. Ann Clin Lab Sci. 2000;30(1):3–21.

    CAS  PubMed  Google Scholar 

  139. Turini M, DuBois R. Cyclooxygenase-2: a therapeutic target. Annu Rev Med. 2002;53:35–57.

    Article  CAS  PubMed  Google Scholar 

  140. Gately S, Li WW. Multiple roles of COX-2 in tumor angiogenesis: a target for antiangiogenic therapy. Semin Oncol. 2004;31:2–11.

    Article  CAS  PubMed  Google Scholar 

  141. Kakiuchi Y, Tsuji S, Tsujii M, et al. Cyclooxygenase-2 activity altered the cell-surface carbohydrate antigens on colon cancer cells and enhanced liver metastasis. Cancer Res. 2002;62(5):1567–72.

    CAS  PubMed  Google Scholar 

  142. Tatsuguchi A, Matsui K, Shinji Y, et al. Cyclooxygenase-2 expression correlates with angiogenesis and apoptosis in gastric cancer tissue. Hum Pathol. 2004;35(4):488–95.

    Article  CAS  PubMed  Google Scholar 

  143. Tsujii M, Kawano S, DuBois RN. Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci U S A. 1997;94(7):3336–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Tsujii M, Kawano S, Tsuji S, Sawaoka H, Hori M, DuBois RN. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell. 1998;93(5):705–16.

    Article  CAS  PubMed  Google Scholar 

  145. Wu C, Wang K, McDyer J, Seder RA. Prostaglandin E2 and dexamethasone inhibit IL-12 receptor expression and IL-12 responsiveness. J Immunol. 1998;161(6):2723–30.

    Article  CAS  PubMed  Google Scholar 

  146. Sharma S, Stolina M, Yang S, et al. Tumor cyclooxygenase 2-dependent suppression of dendritic cell function. Clin Cancer Res. 2003;9(3):961–8.

    CAS  PubMed  Google Scholar 

  147. Harris SG, Padilla J, Koumas L, Ray D, Phipps RP. Prostaglandins as modulators of immunity. Trends Immunol. 2002;23(3):144–50.

    Article  CAS  PubMed  Google Scholar 

  148. Ohno S, Ohno Y, Suzuki N, et al. Multiple roles of cyclooxygenase-2 in endometrial cancer. Anticancer Res. 2005;25:3679–87.

    CAS  PubMed  Google Scholar 

  149. Gou H, Chen X, Zhu J, et al. Expressions of COX-2 and VEGF-C in gastric cancer: correlations with lymphangiogenesis and prognostic implications. J Exp Clin Cancer Res. 2011;30(1):14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Zhang J, Luo J, Ni J, et al. MMP-7 is upregulated by COX-2 and promotes proliferation and invasion of lung adenocarcinoma cells. Eur J Histochem. 2014;58(1):2262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Fukazawa EM, Baiocchi G, Soares FA, et al. Cox-2, EGFR, and ERBB-2 expression in cervical intraepithelial neoplasia and cervical cancer using an automated imaging system. Int J Gynecol Pathol. 2014;33(3):225–34.

    Article  CAS  PubMed  Google Scholar 

  152. Ristimäki A, Sivula A, Lundin J, et al. Prognostic significance of elevated cyclooxygenase-2 expression in breast cancer. Cancer Res. 2002;62(3):632–5.

    PubMed  Google Scholar 

  153. Ogino S, Kirkner GJ, Nosho K, et al. Cyclooxygenase-2 expression is an independent predictor of poor prognosis in colon cancer. Clin Cancer Res. 2008;14(24):8221–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Cao QJ, Einstein MH, Anderson PS, Runowicz CD, Balan R, Jones JG. Expression of COX-2, Ki-67, cyclin D1, and P21 in endometrial endometrioid carcinomas. Int J Gynecol Pathol. 2002;21(2):147–54.

    Article  PubMed  Google Scholar 

  155. Ferrandina G, Legge F, Ranelletti FO, et al. Cyclooxygenase-2 expression in endometrial carcinoma: correlation with clinicopathologic parameters and clinical outcome. Cancer. 2002;95(4):801–7.

    Article  CAS  PubMed  Google Scholar 

  156. Ohno Y, Ohno S, Suzuki N, et al. Role of cyclooxygenase-2 in immunomodulation and prognosis of endometrial carcinoma. Int J Cancer. 2005;114(5):696–701.

    Article  CAS  PubMed  Google Scholar 

  157. Suemori T, Susumu N, Iwata T, et al. Intratumoral CD8+ Lymphocyte Infiltration as a Prognostic Factor and Its Relationship With Cyclooxygenase 2 Expression and Microsatellite Instability in Endometrial Cancer. Int J Gynecol Cancer. 2015;25(7):1165–72.

    Article  PubMed  Google Scholar 

  158. Bell SC, Patel S, Hales MW, Kirwan PH, Drife JO. fertility. Immunochemical detection and characterization of pregnancy-associated endometrial alpha 1- and alpha 2-globulins secreted by human endometrium and decidua. J Reprod Fertil. 1985;74(1):261–70.

    Article  CAS  PubMed  Google Scholar 

  159. Tseng L, Mazella J. Endometrial cell specific gene activation during implantation and early pregnancy. Front Biosci. 2002;7:d1566–74.

    Article  PubMed  Google Scholar 

  160. Hoozemans DA, Schats R, Lambalk CB, Homburg R, Hompes PG. Human embryo implantation: current knowledge and clinical implications in assisted reproductive technology. Reprod Biomed Online. 2004;9(6):692–715.

    Article  PubMed  Google Scholar 

  161. Cui J, Liu YG, Wang XW. The Roles of Glycodelin in Cancer Development and Progression. Front Immunol. 2017;8:1685.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Seppälä M, Koistinen H, Koistinen R, Hautala L, Chiu PC, Yeung WS. Glycodelin in reproductive endocrinology and hormone-related cancer. Eur J Endocrinol. 2009;160(2):121–33.

    Article  PubMed  CAS  Google Scholar 

  163. Lenhard M, Heublein S, Kunert-Keil C, et al. Immunosuppressive Glycodelin A is an independent marker for poor prognosis in endometrial cancer. BMC Cancer. 2013;13:616.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Schneider MA, Granzow M, Warth A, et al. Glycodelin: A New Biomarker with Immunomodulatory Functions in Non-Small Cell Lung Cancer. Clin Cancer Res. 2015;21(15):3529–40.

    Article  CAS  PubMed  Google Scholar 

  165. Jeschke U, Mylonas I, Kunert-Keil C, et al. Immunohistochemistry, glycosylation and immunosuppression of glycodelin in human ovarian cancer. Histochem Cell Biol. 2009;131(2):283–95.

    Article  CAS  PubMed  Google Scholar 

  166. Hu G, Tu W, Yang L, Peng G, Yang L. ARID1A deficiency and immune checkpoint blockade therapy: From mechanisms to clinical application. Cancer Lett. 2020;473:148–55.

    Article  CAS  PubMed  Google Scholar 

  167. Jiang T, Chen X, Su C, Ren S, Zhou CC. ARID1APan-cancer analysis of Alterations as Biomarkers for Immunotherapy Outcomes. J Cancer. 2020;11(4):776–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Bosse T, ter Haar N, Seeber L, et al. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations, TP53 and microsatellite instability in endometrial cancer. Mod Pathol. 2013;26(11):1525–35.

    Article  CAS  PubMed  Google Scholar 

  169. Parikh A, He Y, Hong T, et al. Analysis of DNA Damage Response Gene Alterations and Tumor Mutational Burden Across 17,486 Tubular Gastrointestinal Carcinomas: Implications for Therapy. Oncologist. 2019;24(10):1340–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Shen J, Ju Z, Zhao W, et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med. 2018;24(5):556–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Li L, Li MY, Jiang ZH, Wang XS. ARID1A mutations are associated with increased immune activity in gastrointestinal cancer. Cells. 2019;8(7):678.

  172. Bell DW, Ellenson LH. Molecular Genetics of Endometrial Carcinoma. Annu Rev Pathol. 2019;14:339–67.

    Article  CAS  PubMed  Google Scholar 

  173. Shukla S, Howitt B, Wu C, Konstantinopoulos PA. Predicted neoantigen load in non-hypermutated endometrial cancers: Correlation with outcome and tumor-specific genomic alterations. Gynecol Oncol Rep. 2017;19:42–5.

    Article  PubMed  Google Scholar 

  174. Boussiotis VA. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N Engl J Med. 2016;375(18):1767–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Topalian S, Drake C, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Ott P, Bang Y, Berton-Rigaud D, et al. Safety and Antitumor Activity of Pembrolizumab in Advanced Programmed Death Ligand 1-Positive Endometrial Cancer: Results From the KEYNOTE-028 Study. J Clin Oncol. 2017;35(22):2535–41.

    Article  CAS  PubMed  Google Scholar 

  178. Marabelle A, Le D, Ascierto P, et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol. 2020;38(1):1–10.

    Article  CAS  PubMed  Google Scholar 

  179. Azad N, Gray R, Overman M, et al. Nivolumab Is Effective in Mismatch Repair-Deficient Noncolorectal Cancers: Results From Arm Z1D-A Subprotocol of the NCI-MATCH (EAY131) Study. J Clin Oncol. 2020;38(3):214–22.

    Article  CAS  PubMed  Google Scholar 

  180. Fleming GF, Emens LA, Eder JP, Hamilton, Joyce F. Liu, Bo Liu, Luciana Molinero, Marcella Fasso, Carol O'Hear, Fadi S. Braiteh, (2017) Clinical activity, safety and biomarker results from a phase Ia study of atezolizumab (atezo) in advanced/recurrent endometrial cancer (rEC). J Clin Oncol. 2017;35(15_suppl):5585.

  181. Konstantinopoulos P, Luo W, Liu J, et al. Phase II Study of Avelumab in Patients With Mismatch Repair Deficient and Mismatch Repair Proficient Recurrent/Persistent Endometrial Cancer. J Clin Oncol. 2019;37(30):2786–94.

    Article  CAS  PubMed  Google Scholar 

  182. Antill A, Kok PS, Stockler MR, Robledo K, Yip S, Parry M, Smith D, Spurdle A, Barnes E, Friedlander ML, Baron-Hay S, Shannon C, Coward J, Beale PJ, Goss G, Meniawy T, Andrews J, Kelly M, Mileshkin. Updated results of activity of durvalumab in advanced endometrial cancer (AEC) according to mismatch repair (MMR) status: The phase II PHAEDRA trial (ANZGOG1601). Annals of Oncology. 2019;30:ix192

  183. Calabrò L, Morra A, Giannarelli D, et al. Tremelimumab combined with durvalumab in patients with mesothelioma (NIBIT-MESO-1): an open-label, non-randomised, phase 2 study. Lancet Respir Med. 2018;6(6):451–60.

    Article  PubMed  Google Scholar 

  184. Maio M, Scherpereel A, Calabrò L, et al. Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): a multicentre, international, randomised, double-blind, placebo-controlled phase 2b trial. Lancet Oncol. 2017;18(9):1261–73.

    Article  CAS  PubMed  Google Scholar 

  185. Antonia S, Goldberg S, Balmanoukian A, et al. Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study. Lancet Oncol. 2016;17(3):299–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. MM Rubinstein, Caird I, Zhou Q, Iasonos A, Friedman SF, Cadoo KA, Konner JA, O'Cearbhaill RE, Tew WP, Zamarin D, Grisham RN, Kyi C, Soldan K, Lichtman SM, Ligresti L, Schweber SJ, Singh JC, Troso-Sandoval TA, Aghajanian C, Makker V. A phase II trial of durvalumab with or without tremelimumab in patients with persistent or recurrent endometrial carcinoma and endometrial carcinosarcoma. J Clin Oncol. 2019;37 (15_suppl):5582.

  187. Mahmood S, Chen C, Shapnik N, Krishnan U, Singh H. Makker VJGor. Myocarditis with tremelimumab plus durvalumab combination therapy for endometrial cancer: A case report. Gynecol Oncol Rep. 2018;25:74–7.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Brooks R, Fleming G, Lastra R, et al. Current recommendations and recent progress in endometrial cancer. CA Cancer J Clin. 2019;69(4):258–79.

    PubMed  Google Scholar 

  189. Goodman AM, Sokol ES, Frampton GM, Lippman SM, Kurzrock R. Microsatellite-Stable Tumors with High Mutational Burden Benefit from Immunotherapy. Cancer Immunol Res. 2019;7(10):1570–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Makker V, Rasco D, Vogelzang N, et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer: an interim analysis of a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019;20(5):711–8.

    Article  CAS  PubMed  Google Scholar 

  191. Aghajanian C, Sill MW, Darcy KM, Greer B, McMeekin DS, Peter PG, Rotmensch J, Barnes MN, Hanjani P, Leslie KK, (2011) Phase II Trial of Bevacizumab in Recurrent or Persistent Endometrial Cancer: A Gynecologic Oncology Group Study. J Clin Oncol. 2011;29(16):2259-65.

  192. McMeekin S, Dizon D, Barter J, Scambia G, Manzyuk L, Lisyanskaya A, Oaknin A, Ringuette S, Mukhopadhyay P, Rosenberg J, Vergote I. Phase III randomized trial of second-line ixabepilone versus paclitaxel or doxorubicin in women with advanced endometrial cancer. Gynecol Oncol. 20915;138(1):18–23

  193. Oh MS, Chae. YW. Deep and Durable Response With Combination CTLA-4 and PD-1 Blockade in Mismatch Repair (MMR)-proficient Endometrial Cancer. J Immunother. 2019;42(2):51–4

  194. Chari RVJ. Targeted cancer therapy: conferring specificity to cytotoxic drugs. Acc Chem Res. 2008;41(1):98–107.

    Article  CAS  PubMed  Google Scholar 

  195. Lambert JM. Drug-conjugated antibodies for the treatment of cancer. Br J Clin Pharmacol. 2013;76(2):248–62.

    Article  CAS  PubMed  Google Scholar 

  196. Chari RVJ, Miller ML, Widdison WC. Antibody-drug conjugates: an emerging concept in cancer therapy. Angew Chem Int Ed Engl. 2014;53(15):3796–827.

    Article  CAS  PubMed  Google Scholar 

  197. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367(19):1783–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Moore K, Martin L, O'Malley D, et al. Safety and activity of mirvetuximab soravtansine (IMGN853), a folate receptor alpha-targeting antibody-drug conjugate, in platinum-resistant ovarian, fallopian tube, or primary peritoneal cancer: a phase i expansion study. J Clin Oncol. 2017;35(10):1112–8.

    Article  CAS  PubMed  Google Scholar 

  199. Erickson H, Widdison W, Mayo M, et al. Tumor delivery and in vivo processing of disulfide-linked and thioether-linked antibody-maytansinoid conjugates. Bioconjug Chem. 2010;21(1):84–92.

    Article  CAS  PubMed  Google Scholar 

  200. Lopus M, Oroudjev E, Wilson L, et al. Maytansine and cellular metabolites of antibody-maytansinoid conjugates strongly suppress microtubule dynamics by binding to microtubules. Mol Cancer Ther. 2010;9(10):2689–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Oroudjev E, Lopus M, Wilson L, et al. Maytansinoid-antibody conjugates induce mitotic arrest by suppressing microtubule dynamic instability. Mol Cancer Ther. 2010;9(10):2700–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Shen J, Peng Y, Wei L, et al. ARID1A deficiency impairs the DNA damage checkpoint and sensitizes cells to PARP inhibitors. Cancer Discov. 2015;5(7):752–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Miyasaka A, Oda K, Ikeda Y, et al. Anti-tumor activity of olaparib, a poly (ADP-ribose) polymerase (PARP) inhibitor, in cultured endometrial carcinoma cells. BMC Cancer. 2014;14:179.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Park Y, Chui M, Suryo Rahmanto Y, et al. Loss of ARID1A in tumor cells renders selective vulnerability to combined ionizing radiation and PARP inhibitor therapy. Clin Cancer Res. 2019;25(18):5584–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Triebel F, Jitsukawa S, Baixeras E, et al. LAG-3, a novel lymphocyte activation gene closely related to CD4. J Exp Med. 1990;171(5):1393–405.

    Article  CAS  PubMed  Google Scholar 

  206. Baixeras E, Huard B, Miossec C, et al. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens. J Exp Med. 1992;176(2):327–37.

    Article  CAS  PubMed  Google Scholar 

  207. Huard B, Prigent P, Pagès F, Bruniquel D, Triebel F. T cell major histocompatibility complex class II molecules down-regulate CD4+ T cell clone responses following LAG-3 binding. Eur J Immunol. 1996;26(5):1180–6.

    Article  CAS  PubMed  Google Scholar 

  208. Workman C, Vignali DAA. The CD4-related molecule, LAG-3 (CD223), regulates the expansion of activated T cells. Eur J Immunol. 2003;33(4):970–9.

    Article  CAS  PubMed  Google Scholar 

  209. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity. 2016;44(5):989–1004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Workman C, Cauley L, Kim I, Blackman M, Woodland D. Vignali DJJoi. Lymphocyte activation gene-3 (CD223) regulates the size of the expanding T cell population following antigen activation in vivo. J Immunol. 2004;172(9):5450–5.

    Article  CAS  PubMed  Google Scholar 

  211. Workman C, Rice D, Dugger K, Kurschner C, Vignali DJE. Phenotypic analysis of the murine CD4-related glycoprotein, CD223 (LAG-3). Eur J Immunol. 2002;32(8):2255–63.

    Article  CAS  PubMed  Google Scholar 

  212. Workman C. Vignali DJJoi. Negative regulation of T cell homeostasis by lymphocyte activation gene-3 (CD223). J Immunol. 2005;174(2):688–95.

    Article  CAS  PubMed  Google Scholar 

  213. Wang J, Sanmamed M, Datar I, et al. Fibrinogen-like protein 1 is a major immune inhibitory ligand of LAG-3. Cell. 2019;176:334–347.e12.

    Article  CAS  PubMed  Google Scholar 

  214. Sica G, Choi I, Zhu G, et al. B7-H4, a molecule of the B7 family, negatively regulates T cell immunity. Immunity. 2003;18(6):849–61.

    Article  CAS  PubMed  Google Scholar 

  215. Carreno B, Collins M. BTLA: a new inhibitory receptor with a B7-like ligand. Trends Immunol. 2003;24(10):524–7.

    Article  CAS  PubMed  Google Scholar 

  216. Greenwald R, Freeman G, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–48.

    Article  PubMed  CAS  Google Scholar 

  217. Wang JY, Wang WP. B7-H4, a promising target for immunotherapy. Cell Immunol. 2020;347:104008.

    Article  CAS  PubMed  Google Scholar 

  218. Choi I, Zhu G, Sica G, et al. Genomic organization and expression analysis of B7-H4, an immune inhibitory molecule of the B7 family. J Immunol. 2003;171(9):4650–4.

    Article  CAS  PubMed  Google Scholar 

  219. Fan M, Zhuang Q, Chen Y, et al. B7-H4 expression is correlated with tumor progression and clinical outcome in urothelial cell carcinoma. Int J Clin Exp Pathol. 2014;7(10):6768–75.

    PubMed  PubMed Central  Google Scholar 

  220. Smith JB, Stashwick C, Powell DJ. B7-H4 as a potential target for immunotherapy for gynecologic cancers: a closer look. Gynecol Oncol. 2014;134(1):181–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Schalper K, Carvajal-Hausdorf D, McLaughlin J, et al. Differential Expression and Significance of PD-L1, IDO-1, and B7-H4 in Human Lung Cancer. Clin Cancer Res. 2017;23(2):370–8.

    Article  CAS  PubMed  Google Scholar 

  222. Liang L, Jiang Y, Chen J, et al. B7-H4 expression in ovarian serous carcinoma: a study of 306 cases. Hum Pathol. 2016;57:1–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Xu H, Chen X, Tao M, et al. B7-H3 and B7-H4 are independent predictors of a poor prognosis in patients with pancreatic cancer. Oncol Lett. 2016;11(3):1841–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Liu L, Li D, Chen S, et al. B7-H4 expression in human infiltrating ductal carcinoma-associated macrophages. Mol Med Rep. 2016;14(3):2135–42.

    Article  CAS  PubMed  Google Scholar 

  225. Miyatake T, Tringler B, Liu W, et al. B7-H4 (DD-O110) is overexpressed in high risk uterine endometrioid adenocarcinomas and inversely correlated with tumor T-cell infiltration. Gynecol Oncol. 2007;106(1):119–27.

    Article  CAS  PubMed  Google Scholar 

  226. Liu J, Liu YL, Wang WL, Wang CY, Che YH. medicine t. Expression of immune checkpoint molecules in endometrial carcinoma. Exp Ther Med. 2015;10(5):1947–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Bregar A, Deshpande A, Grange C, et al. Characterization of immune regulatory molecules B7-H4 and PD-L1 in low and high grade endometrial tumors. Gynecol Oncol. 2017;145(3):446–52.

    Article  CAS  PubMed  Google Scholar 

  228. Qin S, Xu LP, Yi M, Yu SN, Wu KM, Luo SX. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer. 2019;18(1):155.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  229. Anderson AC. Tim-3, a negative regulator of anti-tumor immunity. Curr Opin Immunol. 2012;24(2):213–6.

    Article  CAS  PubMed  Google Scholar 

  230. Zhu C, Anderson A, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005;6(12):1245–52.

    Article  CAS  PubMed  Google Scholar 

  231. Du W, Yang M, Turner A, et al. TIM-3 as a Target for Cancer Immunotherapy and Mechanisms of Action. Int J Mol Sci. 2017;18(3):645.

  232. Sánchez-Fueyo A, Tian J, Picarella D, et al. Tim-3 inhibits T helper type 1-mediated auto- and alloimmune responses and promotes immunological tolerance. Nat Immunol. 2003;4(11):1093–101.

    Article  PubMed  Google Scholar 

  233. Rangachari M, Zhu C, Sakuishi K, et al. Bat3 promotes T cell responses and autoimmunity by repressing Tim-3–mediated cell death and exhaustion. Nat Med. 2012;18(9):1394–400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Das M, Zhu C, Kuchroo VK. Tim-3 and its role in regulating anti-tumor immunity. Immunol Rev. 2017;276(1):97–111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Jiang J, Jin M, Kong F, et al. Decreased galectin-9 and increased Tim-3 expression are related to poor prognosis in gastric cancer. PLoS One. 2013;8(12):e81799.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  236. Cao Y, Zhou X, Huang X, et al. Correction: Tim-3 Expression in Cervical Cancer Promotes Tumor Metastasis. PLoS One. 2016;11(3):e0152830.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  237. Yuan J, Jiang B, Zhao H, Huang QJN. Prognostic implication of TIM-3 in clear cell renal cell carcinoma. Neoplasma. 2014;61(1):35–40.

    Article  CAS  PubMed  Google Scholar 

  238. Datar I, Sanmamed M, Wang J, et al. Expression analysis and significance of PD-1, LAG-3, and TIM-3 in human non-small cell lung cancer using spatially resolved and multiparametric single-cell analysis. Clin Cancer Res. 2019;25(15):4663–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Moore M, Ring KL, Mills AM. Canadian academy of pathology I. TIM-3 in endometrial carcinomas: an immunotherapeutic target expressed by mismatch repair-deficient and intact cancers. Mod Pathol. 2019;32(8):1168–79.

    Article  CAS  PubMed  Google Scholar 

  240. Silk AW, Finn OJ. Cancer vaccines: a promising cancer therapy against all odds. Future Oncol. 2007;3(3):299–306.

    Article  CAS  PubMed  Google Scholar 

  241. Emens LA. Cancer vaccines: on the threshold of success. Expert Opin Emerg Drugs. 2008;13(2):295–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Hu ZT, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 2018;18(3):168–82.

    Article  CAS  PubMed  Google Scholar 

  243. Bocchia M, Bronte V, Colombo M, et al. Antitumor vaccination: where we stand. Haematologica. 2000;85(11):1172–206.

    CAS  PubMed  Google Scholar 

  244. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  CAS  PubMed  Google Scholar 

  245. Van Lint S, Wilgenhof S, Heirman C, et al. Optimized dendritic cell-based immunotherapy for melanoma: the TriMix-formula. Cancer Immunol Immunother. 2014;63(9):959–67.

    Article  PubMed  CAS  Google Scholar 

  246. Giarelli E. Cancer vaccines: a new frontier in prevention and treatment. Oncology (Williston Park). 2007;21:11–7 discussion 18.

    PubMed  Google Scholar 

  247. Guo C, Manjili M, Subjeck J, Sarkar D, Fisher P, Wang XY. Therapeutic cancer vaccines: past, present, and future. Adv Cancer Res. 2013;119:421–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Yang L, Han Y, Suarez Saiz F, Minden MD. A tumor suppressor and oncogene: the WT1 story. Leukemia. 2007;21(5):868–76.

    Article  CAS  PubMed  Google Scholar 

  249. Nakatsuka S, Oji Y, Horiuchi T, et al. Immunohistochemical detection of WT1 protein in a variety of cancer cells. Mod Pathol. 2006;19(6):804–14.

    Article  CAS  PubMed  Google Scholar 

  250. Ohno S, Dohi S, Ohno Y, et al. Immunohistochemical detection of WT1 protein in endometrial cancer. Anticancer Res. 2009;29(5):1691–5.

    PubMed  Google Scholar 

  251. Coosemans A, Wölfl M, Berneman Z, et al. Immunological response after therapeutic vaccination with WT1 mRNA-loaded dendritic cells in end-stage endometrial carcinoma. Anticancer Res. 2010;30(9):3709–14.

    PubMed  Google Scholar 

  252. Miyatake T, Ueda Y, Morimoto A, et al. WT1 peptide immunotherapy for gynecologic malignancies resistant to conventional therapies: a phase II trial. J Cancer Res Clin Oncol. 2013;139(3):457–63.

    Article  CAS  PubMed  Google Scholar 

  253. Coosemans A, Vanderstraeten A, Tuyaerts S, et al. Wilms' Tumor Gene 1 (WT1)--loaded dendritic cell immunotherapy in patients with uterine tumors: a phase I/II clinical trial. Anticancer Res. 2013;33(12):5495–500.

    CAS  PubMed  Google Scholar 

  254. Ohno S, Kyo S, Myojo S, et al. Wilms' tumor 1 (WT1) peptide immunotherapy for gynecological malignancy. Anticancer Res. 2009;29(11):4779–84.

    CAS  PubMed  Google Scholar 

  255. Dranoff G, Jaffee E, Lazenby A, et al. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci U S A. 1993;90(8):3539–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Li P, Del Vecchio S, Fonti R, et al. Local concentration of folate binding protein GP38 in sections of human ovarian carcinoma by in vitro quantitative autoradiography. J Nucl Med. 1996;37(4):665–72.

    CAS  PubMed  Google Scholar 

  257. Wu M, Gunning W, Ratnam M. Expression of folate receptor type alpha in relation to cell type, malignancy, and differentiation in ovary, uterus, and cervix. Cancer Epidemiol Biomarkers Prev. 1999;8(9):775–82.

    CAS  PubMed  Google Scholar 

  258. Brown T, Byrd K, Vreeland T, et al. Final analysis of a phase I/IIa trial of the folate-binding protein-derived E39 peptide vaccine to prevent recurrence in ovarian and endometrial cancer patients. Cancer Med. 2019;8(10):4678–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Chen Y, Scanlan M, Sahin U, et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A. 1997;94(5):1914–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Jungbluth A, Chen Y, Stockert E, et al. Immunohistochemical analysis of NY-ESO-1 antigen expression in normal and malignant human tissues. Int J Cancer. 2001;92(6):856–60.

    Article  CAS  PubMed  Google Scholar 

  261. Scanlan M, Gure A, Jungbluth A, Old L. Chen YJIr. Cancer/testis antigens: an expanding family of targets for cancer immunotherapy. Immunol Rev. 2002;188:22–32.

    Article  CAS  PubMed  Google Scholar 

  262. Jäger E, Karbach J, Gnjatic S, et al. Recombinant vaccinia/fowlpox NY-ESO-1 vaccines induce both humoral and cellular NY-ESO-1-specific immune responses in cancer patients. Proc Natl Acad Sci U S A. 2006;103(39):14453–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  263. Kaumaya P, Foy K, Garrett J, et al. Phase I active immunotherapy with combination of two chimeric, human epidermal growth factor receptor 2, B-cell epitopes fused to a promiscuous T-cell epitope in patients with metastatic and/or recurrent solid tumors. J Clin Oncol. 2009;27(31):5270–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Bekaii-Saab T, Wesolowski R, Ahn D, et al. Phase I Immunotherapy Trial with Two Chimeric HER-2 B-Cell Peptide Vaccines Emulsified in Montanide ISA 720VG and Nor-MDP Adjuvant in Patients with Advanced Solid Tumors. Clin Cancer Res. 2019;25(12):3495–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Schwitalle Y, Kloor M, Eiermann S, et al. Immune response against frameshift-induced neopeptides in HNPCC patients and healthy HNPCC mutation carriers. Gastroenterology. 2008;134(4):988–97.

    Article  CAS  PubMed  Google Scholar 

  266. Yang G, Zheng RY, Jin ZS. Correlations between microsatellite instability and the biological behaviour of tumours. J Cancer Res Clin Oncol. 2019;145(12):2891–9.

    Article  PubMed  PubMed Central  Google Scholar 

  267. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8(4):299–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Fan J, Shang D, Han B, Song J, Chen H, Yang JM. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Theranostics. 2018;8(20):5784–800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Ruella M, Kalos M. Adoptive immunotherapy for cancer. Immunol Rev. 2014;257(1):14–38.

    Article  PubMed  Google Scholar 

  270. Rosenberg S, Lotze M, Muul L, et al. Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med. 1985;313(23):1485–92.

    Article  CAS  PubMed  Google Scholar 

  271. Takei F. LAK cell therapy of AML: not to be lost in translation. Exp Hematol. 2011;39(11):1045–6.

    Article  CAS  PubMed  Google Scholar 

  272. Baxevanis C, Dedoussis G, Papadopoulos N, et al. Enhanced human lymphokine-activated killer cell function after brief exposure to granulocyte-macrophage-colony stimulating factor. Cancer. 1995;76(7):1253–60.

    Article  CAS  PubMed  Google Scholar 

  273. Shimizu H, Inoue M, Tanizawa O. Adoptive cellular immunotherapy to the endometrial carcinoma cell line xenografts in nude mice. Gynecol Oncol. 1989;34(2):195–9.

    Article  CAS  PubMed  Google Scholar 

  274. Steis R, Urba W, VanderMolen L, et al. Intraperitoneal lymphokine-activated killer-cell and interleukin-2 therapy for malignancies limited to the peritoneal cavity. J Clin Oncol. 1990;8(10):1618–29.

    Article  CAS  PubMed  Google Scholar 

  275. Santin A, Hermonat P, Ravaggi A, et al. Development and therapeutic effect of adoptively transferred T cells primed by tumor lysate-pulsed autologous dendritic cells in a patient with metastatic endometrial cancer. Gynecol Obstet Invest. 2000;49(3):194–203.

    Article  CAS  PubMed  Google Scholar 

  276. Sermer D, Brentjens RJH. CAR T-cell therapy: full speed ahead. Hematol Oncol. 2019;37 Suppl 1:95-100.

  277. Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity. 2013;39(1):49–60.

    Article  CAS  PubMed  Google Scholar 

  278. Boyiadzis M, Dhodapkar M, Brentjens R, et al. Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. J Immunother Cancer. 2018;6(1):137.

    Article  PubMed  PubMed Central  Google Scholar 

  279. Pan J, Yang J, Deng B, et al. High efficacy and safety of low-dose CD19-directed CAR-T cell therapy in 51 refractory or relapsed B acute lymphoblastic leukemia patients. Leukemia. 2017;31(12):2587–93.

    Article  CAS  PubMed  Google Scholar 

  280. Sánchez-Martínez D, Baroni M, Gutierrez-Agüera F, et al. Fratricide-resistant CD1a-specific CAR T cells for the treatment of cortical T-cell acute lymphoblastic leukemia. Blood. 2019;133(21):2291–304.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  281. Chow VA, Shadman M, Gopal AK. Translating anti-CD19 CAR T-cell therapy into clinical practice for relapsed/refractory diffuse large B-cell lymphoma. Blood. 2018;132(8):777–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  282. Rodriguez-Garcia A, Sharma P, Poussin M, et al. CAR T Cells Targeting MISIIR for the Treatment of Ovarian Cancer and Other Gynecologic Malignancies. Mol Ther. 2020;28(2):548–60.

    Article  CAS  PubMed  Google Scholar 

  283. Masiakos P, MacLaughlin D, Maheswaran S, et al. Human ovarian cancer, cell lines, and primary ascites cells express the human Mullerian inhibiting substance (MIS) type II receptor, bind, and are responsive to MIS. Clin Cancer Res. 1999;5(11):3488–99.

    CAS  PubMed  Google Scholar 

  284. Renaud EJ, MacLaughlin DT, Oliva E, Rueda BR, Donahoe PK. Endometrial cancer is a receptor-mediated target for Mullerian Inhibiting Substance. Proc Natl Acad Sci U S A. 2005;102(1):111–6.

    Article  CAS  PubMed  Google Scholar 

  285. Bakkum-Gamez J, Aletti G, Lewis K, et al. Müllerian inhibiting substance type II receptor (MISIIR): a novel, tissue-specific target expressed by gynecologic cancers. Gynecol Oncol. 2008;108(1):141–8.

    Article  CAS  PubMed  Google Scholar 

  286. Song J, Chen K, Kim S, et al. The expression of Müllerian inhibiting substance/anti-Müllerian hormone type II receptor protein and mRNA in benign, borderline and malignant ovarian neoplasia. Int J Cancer. 2009;34(6):1583–91.

    CAS  Google Scholar 

  287. Barbie TU, Barbie DA, MacLaughlin DT, Maheswaran S, Donahoe PK. Mullerian Inhibiting Substance inhibits cervical cancer cell growth via a pathway involving p130 and p107. Proc Natl Acad Sci U S A. 2003;100(26):15601–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Pépin D, Sosulski A, Zhang L, et al. AAV9 delivering a modified human Mullerian inhibiting substance as a gene therapy in patient-derived xenografts of ovarian cancer. Proc Natl Acad Sci U S A. 2015;112(32):E4418–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  289. Roti Roti JL. Cellular responses to hyperthermia (40-46 degrees C): cell killing and molecular events. Int J Hyperthermia. 2008;24(1):3–15.

    Article  PubMed  CAS  Google Scholar 

  290. Prat A, Adamo B, Cheang MCU, Anders CK, Carey LA, Perou CM. Molecular characterization of basal-like and non-basal-like triple-negative breast cancer. Oncologist. 2013;18(2):123–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Prat A, Lluch A, Albanell J, et al. Predicting response and survival in chemotherapy-treated triple-negative breast cancer. Br J Cancer. 2014;111(8):1532–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Lheureux S, McCourt C, Rimel B, et al. Moving forward with actionable therapeutic targets and opportunities in endometrial cancer: A NCI clinical trials planning meeting report. Gynecol Oncol. 2018; S0090-8258(18)30124-0

  293. Odunsi K. Immunotherapy in ovarian cancer. Ann Oncol. 2017;28(suppl_8):viii1–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  294. Park JH, Ahn JH, Kim SB. How shall we treat early triple-negative breast cancer (TNBC): from the current standard to upcoming immuno-molecular strategies. ESMO Open. 2018;3(Suppl 1):e000357.

    Article  PubMed  PubMed Central  Google Scholar 

  295. Bianchini G, Balko JM, Mayer IA, Sanders ME, Gianni L. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13(11):674–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Network CGAR. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474(7353):609–15.

    Article  CAS  Google Scholar 

  297. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–4.

    Article  PubMed  Google Scholar 

  298. Horiuchi D, Kusdra L, Huskey N, et al. MYC pathway activation in triple-negative breast cancer is synthetic lethal with CDK inhibition. J Exp Med. 2012;209(4):679–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Kim S, Park S, Cho MS, Lim W, Moon BI. Sung SH Strong Correlation of Indoleamine 2,3-Dioxygenase 1 Expression with Basal-Like Phenotype and Increased Lymphocytic Infiltration in Triple-Negative Breast Cancer. J Cancer. 2017;8(1):124–30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  300. Mills A, Peres L, Meiss A, et al. Targetable immune regulatory molecule expression in high-grade serous ovarian carcinomas in African American women: a study of PD-L1 and IDO in 112 cases from the African American cancer epidemiology study (AACES). Int J Clin Gynecol Pathol. 2019;38(2):157–70.

    Article  CAS  Google Scholar 

  301. Santin A, Bellone S, Van Stedum S, et al. Amplification of c-erbB2 oncogene: a major prognostic indicator in uterine serous papillary carcinoma. Cancer. 2005;104(7):1391–7.

    Article  CAS  PubMed  Google Scholar 

  302. Slomovitz B, Broaddus R, Burke T, et al. Her-2/neu overexpression and amplification in uterine papillary serous carcinoma. J Clin Oncol. 2004;22(15):3126–32.

    Article  CAS  PubMed  Google Scholar 

  303. Grushko T, Filiaci V, Mundt A, et al. An exploratory analysis of HER-2 amplification and overexpression in advanced endometrial carcinoma: a gynecologic oncology group study. Gynecol Oncol. 2008;108(1):3–9.

    Article  CAS  PubMed  Google Scholar 

  304. Díaz-Montes T, Ji H, Smith Sehdev A, et al. Clinical significance of Her-2/neu overexpression in uterine serous carcinoma. Gynecol Oncol. 2006;100(1):139–44.

    Article  PubMed  CAS  Google Scholar 

  305. Roque D, Santin AD. Updates in therapy for uterine serous carcinoma. Opin Obstet Gynecol. 2013;25(1):29–37.

    Article  Google Scholar 

  306. Arnould L, Gelly M, Penault-Llorca F, et al. Trastuzumab-based treatment of HER2-positive breast cancer: an antibody-dependent cellular cytotoxicity mechanism? Br J Cancer. 2006;94(2):259–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  307. Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat Med. 2000;6(4):443–6.

    Article  CAS  PubMed  Google Scholar 

  308. Gennari R, Menard S, Fagnoni F, et al. Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2. Clin Cancer Res. 2004;10(17):5650–5.

    Article  CAS  PubMed  Google Scholar 

  309. Fleming G, Sill M, Darcy K, et al. Phase II trial of trastuzumab in women with advanced or recurrent, HER2-positive endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2010;116(1):15–20.

    Article  CAS  PubMed  Google Scholar 

  310. Fader A, Roque D, Siegel E, et al. Randomized phase II trial of carboplatin-paclitaxel versus carboplatin-paclitaxel-trastuzumab in uterine serous carcinomas that overexpress human epidermal growth factor receptor 2/neu. J Clin Oncol. 2018;36(20):2044–51.

    Article  CAS  PubMed  Google Scholar 

  311. Wickramasinghe D. Tumor and T cell engagement by BiTE. Discov Med. 2013;16(88):149–52.

    PubMed  Google Scholar 

  312. Offner S, Hofmeister R, Romaniuk A, Kufer P, Baeuerle PA. Induction of regular cytolytic T cell synapses by bispecific single-chain antibody constructs on MHC class I-negative tumor cells. Mol Immunol. 2006;43(6):763–71.

    Article  CAS  PubMed  Google Scholar 

  313. Wolf E, Hofmeister R, Kufer P, Schlereth B, Baeuerle PA. BiTEs: bispecific antibody constructs with unique anti-tumor activity. Drug Discov Today. 2005;10(18):1237–44.

    Article  CAS  PubMed  Google Scholar 

  314. Bellone S, Black J, English D, et al. Solitomab, an EpCAM/CD3 bispecific antibody construct (BiTE), is highly active against primary uterine serous papillary carcinoma cell lines in vitro. Am J Obstet Gynecol. 2016;214(1):99.e1–8.

    Article  CAS  Google Scholar 

  315. Schlereth B, Fichtner I, Lorenczewski G, et al. Eradication of tumors from a human colon cancer cell line and from ovarian cancer metastases in immunodeficient mice by a single-chain Ep-CAM-/CD3-bispecific antibody construct. Cancer Res. 2005;65(7):2882–9.

    Article  CAS  PubMed  Google Scholar 

  316. English D, Bellone S, Schwab C, et al. Solitomab, an epithelial cell adhesion molecule/CD3 bispecific antibody (BiTE), is highly active against primary chemotherapy-resistant ovarian cancer cell lines in vitro and fresh tumor cells ex vivo. Cancer. 2015;121(3):403–12.

    Article  CAS  PubMed  Google Scholar 

  317. Grundy SM. Metabolic syndrome: connecting and reconciling cardiovascular and diabetes worlds. J Am Coll Cardiol. 2006;47(6):1093–100.

    Article  CAS  PubMed  Google Scholar 

  318. Yang X, Wang JL. The Role of Metabolic Syndrome in Endometrial Cancer: A Review. Front Oncol. 2019;9:744.

    Article  PubMed  PubMed Central  Google Scholar 

  319. Iyengar NM, Gucalp A, Dannenberg AJ, Hudis CA. Obesity and Cancer Mechanisms: Tumor Microenvironment and Inflammation. J Clin Oncol. 2016;34(35):4270–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  320. Sahoo SS, Zhang XD, Hondermarck H, Tanwar PS. The emerging role of the microenvironment in endometrial cancer. Cancers (Basel). 2018;10(11):408.

  321. de Visser K, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer. 2006;6(1):24–37.

    Article  PubMed  CAS  Google Scholar 

  322. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  323. Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell. 2005;7(3):211–7.

    Article  CAS  PubMed  Google Scholar 

  324. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol. 2011;11(2):85–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  325. Kolb R, Kluz P, Tan Z, et al. Obesity-associated inflammation promotes angiogenesis and breast cancer via angiopoietin-like 4. Oncogene. 2019;38(13):2351–63.

    Article  CAS  PubMed  Google Scholar 

  326. Nieman KM, Romero IL, Van Houten BV, Lengyel E. Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim Biophys Acta. 2013;1831(10):1533–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  327. Bohn T, Rapp S, Luther N, et al. Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat Immunol. 2018;19(12):1319–29.

    Article  CAS  PubMed  Google Scholar 

  328. Hursting S, Hursting MJ. Growth signals, inflammation, and vascular perturbations: mechanistic links between obesity, metabolic syndrome, and cancer. Arterioscler Thromb Vasc Biol. 2012;32(8):1766–70.

    Article  CAS  PubMed  Google Scholar 

  329. Ye JP. Mechanisms of insulin resistance in obesity. Front Med. 2013;7(1):14–24.

    Article  PubMed  PubMed Central  Google Scholar 

  330. Szeto H, Liu S, Soong Y, Alam N, Prusky GT, Seshan SV. Protection of mitochondria prevents high-fat diet-induced glomerulopathy and proximal tubular injury. Kidney Int. 2016;90(5):997–1011.

    Article  CAS  PubMed  Google Scholar 

  331. Mu N, Zhu YX, Wang YM, Zhang HY, Xue FX. Insulin resistance: a significant risk factor of endometrial cancer. Gynecol Oncol. 2012;125(3):751–7.

    Article  CAS  PubMed  Google Scholar 

  332. Kheirandish M, Mahboobi H, Yazdanparast M, Kamal MA. Challenges Related to Glycemic Control in Type 2 Diabetes Mellitus Patients. Curr Drug Metab. 2017;18(2):157–62.

    Article  CAS  PubMed  Google Scholar 

  333. Zhao Y, Sun H, Feng M, et al. Metformin is associated with reduced cell proliferation in human endometrial cancer by inbibiting PI3K/AKT/mTOR signaling. Gynecol Endocrinol. 2018;34(5):428–32.

    Article  CAS  PubMed  Google Scholar 

  334. Kheirandish M, Mahboobi H, Yazdanparast M, Kamal W, Kamal MA. Anti-cancer Effects of Metformin: Recent Evidences for its Role in Prevention and Treatment of Cancer. Curr Drug Metab. 2018;19(9):793–7.

    Article  CAS  PubMed  Google Scholar 

  335. Meireles C, Pereira S, Valadares L, et al. Effects of metformin on endometrial cancer: Systematic review and meta-analysis. Gynecol Oncol. 2017;147(1):167–80.

    Article  CAS  PubMed  Google Scholar 

  336. Sivalingam V, Kitson S, McVey R, et al. Measuring the biological effect of presurgical metformin treatment in endometrial cancer. Br J Cancer. 2016;114(3):281–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  337. Kim J, Lee J, Kim C, Choi J, Kim A. Anti-cancer effect of metformin by suppressing signaling pathway of HER2 and HER3 in tamoxifen-resistant breast cancer cells. Tumour Biol. 2016;37(5):5811–9.

    Article  CAS  PubMed  Google Scholar 

  338. Hart T, Dider S, Han WW, Xu H, Zhao ZM, Xie L. Toward Repurposing Metformin as a Precision Anti-Cancer Therapy Using Structural Systems Pharmacology. Sci Rep. 2016;6:20441.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  339. Schrauwen S, Depreeuw J, Coenegrachts L, Hermans E, Lambrechts D, Amant F. Dual blockade of PI3K/AKT/mTOR (NVP-BEZ235) and Ras/Raf/MEK (AZD6244) pathways synergistically inhibit growth of primary endometrioid endometrial carcinoma cultures, whereas NVP-BEZ235 reduces tumor growth in the corresponding xenograft models. Gynecol Oncol. 2015;138(1):165–73.

    Article  CAS  PubMed  Google Scholar 

  340. Cha J, Yang W, Xia W, et al. Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Mol Cell. 2018;71(4):606–620.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  341. Eikawa S, Nishida M, Mizukami S, Yamazaki C, Nakayama E, Udono H. Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci U S A. 2015;112(6):1809–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  342. Kim JJ, Kurita T, Bulun SE. Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr Rev. 2013;34(1):130–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  343. Kamal A, Tempest N, Parkes C, et al. Hormones and endometrial carcinogenesis. Horm Mol Biol Clin Investig. 2016;25(2):129–48.

    CAS  PubMed  Google Scholar 

  344. Hapangama DK, Kamal AM, Bulmer JN. Estrogen receptor β: the guardian of the endometrium. Hum Reprod Update. 2015;21(2):174–93.

    Article  CAS  PubMed  Google Scholar 

  345. Kurita T, Wang Y, Donjacour A, et al. Paracrine regulation of apoptosis by steroid hormones in the male and female reproductive system. Cell Death Differ. 2001;8(2):192–200.

    Article  CAS  PubMed  Google Scholar 

  346. Li Q, Kannan A, DeMayo F, et al. The antiproliferative action of progesterone in uterine epithelium is mediated by Hand2. Science. 2011;331(6019):912–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  347. Mathew D, Drury JA, Valentijn AJ, Vasieva O, Hapangama DK. In silico, in vitro and in vivo analysis identifies a potential role for steroid hormone regulation of FOXD3 in endometriosis-associated genes. Hum Reprod. 2016;31(2):345–54.

    CAS  PubMed  Google Scholar 

  348. Pierdominici M, Maselli A, Colasanti T, et al. Estrogen receptor profiles in human peripheral blood lymphocytes. Immunol Lett. 2010;132:79–85.

  349. Kovats S. Estrogen receptors regulate innate immune cells and signaling pathways. Cell Immunol. 2015;294(2):63–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  350. Mantalaris A, Panoskaltsis N, Sakai Y, et al. Localization of androgen receptor expression in human bone marrow. J Pathol. 2001;193(3):361–6.

    Article  CAS  PubMed  Google Scholar 

  351. Arruvito L, Giulianelli S, Flores A, et al. NK cells expressing a progesterone receptor are susceptible to progesterone-induced apoptosis. J Immunol. 2008;180(8):5746–53.

    Article  CAS  PubMed  Google Scholar 

  352. Özdemir BC, Dotto GP. Sex Hormones and Anticancer Immunity. Clin Cancer Res. 2019;25(15):4603–10.

    Article  PubMed  Google Scholar 

  353. Straub RH. The complex role of estrogens in inflammation. Endocr Rev. 2007;28(5):521–74.

    Article  CAS  PubMed  Google Scholar 

  354. Tanriverdi F, Silveira LFG, MacColl GS, Bouloux PMG. The hypothalamic-pituitary-gonadal axis: immune function and autoimmunity. J Endocrinol. 2003;176(3):293–304.

    Article  CAS  PubMed  Google Scholar 

  355. Cohen-Solal JFG, Jeganathan V, Grimaldi CM, Peeva E, Diamond B. Sex hormones and SLE: influencing the fate of autoreactive B cells. Curr Top Microbiol Immunol. 2006;305:67–88.

    CAS  PubMed  Google Scholar 

  356. Canellada A, Blois S, Gentile T, Margni Idehu RA. In vitro modulation of protective antibody responses by estrogen, progesterone and interleukin-6. Am J Reprod Immunol. 2002;48(5):334–43.

    Article  PubMed  Google Scholar 

  357. De León-Nava M, Nava K, Soldevila G, et al. Immune sexual dimorphism: effect of gonadal steroids on the expression of cytokines, sex steroid receptors, and lymphocyte proliferation. J Steroid Biochem Mol Biol. 2009;113:57–64.

    Article  PubMed  CAS  Google Scholar 

  358. Piccinni M, Giudizi M, Biagiotti R, et al. Progesterone favors the development of human T helper cells producing Th2-type cytokines and promotes both IL-4 production and membrane CD30 expression in established Th1 cell clones. J Immunol. 1995;155(1):128–33.

    Article  CAS  PubMed  Google Scholar 

  359. Witkiewicz AK, McConnell T, Potoczek M, Emmons RVB, Kurman RJ. Increased natural killer cells and decreased regulatory T cells are seen in complex atypical endometrial hyperplasia and well-differentiated carcinoma treated with progestins. Hum Pathol. 2010;41(1):26–32.

    Article  CAS  PubMed  Google Scholar 

  360. Trovik J, Wik E, Werner H, et al. Hormone receptor loss in endometrial carcinoma curettage predicts lymph node metastasis and poor outcome in prospective multicentre trial. Eur J Cancer. 2013;49(16):3431–41.

    Article  CAS  PubMed  Google Scholar 

  361. Ballester M, Canlorbe G, Cortez A, et al. Histological and immunohistochemical profiles predict lymph node status in women with low-intermediate risk endometrial cancer. Gynecol Oncol. 2013;130(3):457–62.

    Article  PubMed  Google Scholar 

  362. Huvila J, Talve L, Carpén O, et al. Progesterone receptor negativity is an independent risk factor for relapse in patients with early stage endometrioid endometrial adenocarcinoma. Gynecol Oncol. 2013;130(3):463–9.

    Article  CAS  PubMed  Google Scholar 

  363. Köbel M, Atenafu E, Rambau P, et al. Progesterone receptor expression is associated with longer overall survival within high-grade histotypes of endometrial carcinoma: A Canadian high risk endometrial cancer consortium (CHREC) study. Gynecol Oncol. 2016;141(3):559–63.

    Article  PubMed  CAS  Google Scholar 

  364. Karnezis A, Leung S, Magrill J, et al. Evaluation of endometrial carcinoma prognostic immunohistochemistry markers in the context of molecular classification. J Pathol Clin Res. 2017;3(4):279–93.

  365. Stelloo E, Nout R, Osse E, et al. Improved Risk Assessment by Integrating Molecular and Clinicopathological Factors in Early-stage Endometrial Cancer-Combined Analysis of the PORTEC Cohorts. Clin Cancer Res. 2016;22(16):4215–24.

    Article  CAS  PubMed  Google Scholar 

  366. Jiang X, Tang Q, Li H, et al. Tumor-associated macrophages correlate with progesterone receptor loss in endometrial endometrioid adenocarcinoma. J Obstet Gynaecol Res. 2013;39(4):855–63.

    Article  PubMed  CAS  Google Scholar 

  367. Giatromanolaki A, Bates G, Koukourakis M, et al. The presence of tumor-infiltrating FOXP3+ lymphocytes correlates with intratumoral angiogenesis in endometrial cancer. Gynecol Oncol. 2008;110(2):216–21.

    Article  CAS  PubMed  Google Scholar 

  368. Jerzak K, Duska L. MacKay HJGo. Endocrine therapy in endometrial cancer: An old dog with new tricks. Gynecol Oncol. 2019;153(1):175–83.

    Article  CAS  PubMed  Google Scholar 

  369. Coley WB. II. Contribution to the Knowledge of Sarcoma. Ann Surg. 1891;14(3):199-220.

Download references

Acknowledgements

We sincerely thank all participants in the study.

Funding

This work was supported by the National Natural Science Foundation of China (81874107), Special Foundation of China Postdoctoral Science (2019T120595) and General Program of China Postdoctoral Science Foundation (2019M652402).

Author information

Authors and Affiliations

Authors

Contributions

This study was conceived, designed and interpreted by ZQ . CWY and MXY drafted the manuscript and collected the related references. Fischer JV. contributed to the revision of final manuscript. SCG participated in discussion. KBH designed this review and inspected the manuscript. All authors contributed to this manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Qing Zhang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cao, W., Ma, X., Fischer, J.V. et al. Immunotherapy in endometrial cancer: rationale, practice and perspectives. Biomark Res 9, 49 (2021). https://doi.org/10.1186/s40364-021-00301-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-021-00301-z

Keywords