Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2022 Feb 15;12(2):681–694.

The expression of cancer-testis antigen in ovarian cancer and the development of immunotherapy

Jianhang Zhao 1,*, Zhaoxu Xu 1,*, Yan Liu 2, Xiaobin Wang 3, Xinli Liu 4, Yanan Gao 1, Ying Jin 2
PMCID: PMC8899981  PMID: 35261795

Abstract

Ovarian cancer is a relatively common tumor in women with the highest mortality among female reproductive system tumors. The lack of apparent early symptoms and effective screening strategies often leads to ovarian cancer being diagnosed at an advanced stage. Immunotherapy relying on tumor-associated antigens might improve the treatment of ovarian cancer. Cancer-testis antigens (CTAs) are ideal tumor-associated antigens, and MAGE-A, NY-ESO-1, CT45, and Sp17 are classic CTAs highly expressed in ovarian cancer. Here, we review the research on CTAs in ovarian cancer, including prognostic value and advances in immunotherapy, all of which are essential for developing a theoretical basis for targeted therapy strategies.

Keywords: Cancer-testis antigen, ovarian cancer, immunotherapy, MAGE-A/NY-ESO-1/CT45/Sp17

Introduction

Ovarian cancer is the seventh most common cancer among women and the eighth-most common cause of cancer death worldwide [1]. According to the latest model-based estimates issued by Cancer Statistics in the US, the estimated number of new ovarian cancer patients in 2020 was 21,750, and the death toll was estimated to be 13,940 [2]. Currently, treatment options for ovarian cancer are limited to surgery, radiotherapy, and chemotherapy. Due to the difficulty of achieving an early diagnosis, postoperative tumor recurrence, and late chemotherapy resistance, the 5-year survival rate of advanced ovarian cancer is less than 30% [3]. There is an urgent need for new treatment methods to prolong survival for these reasons.

Tumor immunotherapy restores normal anti-tumor immune responses by restarting and maintaining the tumor-immune cycle, thereby controlling and eliminating tumors [4]. Cancer vaccines rely on tumor antigens and use human-specific immune cells to recognize malignant cells with tumor antigens [5]. Immunotherapy targeting tumor antigens has emerged as an ideal option for immunotherapy in ovarian cancer. The effect of immunotherapy targeting tumor antigens depends on the high expression of these antigens and the specific responses of T lymphocytes to tumor antigens; suitable tumor antigens need to have these two characteristics [6]. Among the tumor antigens, the cancer-testis antigen (CTA) has significant immunogenicity and unique expression patterns in humans [4]. CTA is typically expressed in normal human testes, and a small amount of expression is also present in early developing embryos, placenta, and ovaries [7]. Several studies showed that CTA is overexpressed in various tumor types and is associated with tumor progression [7]. Because the testis is an immune-exempt area with low expression of human leukocyte antigen molecules, the specific expression patterns of CTAs suggest that they are ideal targets for tumor immunotherapy [8]. The development of CTA-based cancer-specific immunotherapy has been going on for many years, and some classic targets have been studied in-depth, including melanoma antigen family A (MAGE-A) [9], New York esophageal squamous cell carcinoma 1 (NY-ESO-1) [10], CT45 [11], and human sperm protein 17 (Sp17) [12]. In recent years, some CTAs associated with ovarian cancer have been reported, including TEX19 [13], POTE [14,15], and HSP70-2 [16]. Many medications based on these targets are studied in clinical trials (Table 1). Effector T cells that specifically respond to CTA have been detected in patients with significantly improved survival [17,18]. These results suggest the effectiveness of CTA-based tumor immunotherapy. This review aims to summarize the characteristics of the classic CTAs and discuss their expression in ovarian cancer, their prognostic value, and research progress in immunotherapy strategies.

Table 1.

Summary of clinical trials involving CTA that are completed or active

NCT NUMBER INTERVENTION PHASE LAST UPDATE POSTED SPONSORS
NCT03159585 NY-ESO-1-specific TCR Affinity Enhancing Specific T Cell Therapy I 2020 Zhujiang Hospital
Xiangxue Pharmaceutical (and more)
NCT01536054 ALVAC(2)-NY-ESO-1(M)/TRICOM vaccine I 2020 Roswell Park Cancer Institute
NCT02650986 Autologous NY-ESO-1 TCR/dnTGFbetaRII transgenic T cells I/II 2020 Roswell Park Cancer Institute
NCT03691376 Autologous NY-ESO-1-specific CD8-positive T Lymphocytes I 2020 Roswell Park Cancer Institute, Buffalo, New York, United States
NCT02166905 DEC-205/NY-ESO-1 Fusion Protein I/II 2020 Roswell Park Cancer Institute
NCT03017131 Genetically Engineered NY-ESO-1-specific T Lymphocytes I 2020 Roswell Park Cancer Institute
NCT02042430 Epacadostat + cancer-testis antigen 1B Early I 2020 National Cancer Institute (NCI)
NCT01567891 gene-modified T cells I/II 2019 Adaptimmune
NCT00803569 ALVAC(2)-NY-ESO-1(M)/TRICOM vaccine I 2019 Ludwig Institute for Cancer Research
NCT00112957 Recombinant Vaccinia-NY-ESO-1 and Recombinant Fowlpox-NY-ESO-1 II 2018 Ludwig Institute for Cancer Research
NCT00616941 NY-ESO-1 OLP4 peptide vaccine I 2018 Ludwig Institute for Cancer Research
NCT03132922 Autologous genetically modified MAGE-A4ᶜ¹º³²T cells I 2017 Adaptimmune
NCT02015416 recombinant NY-ESO-1 antigen and the adjuvant GLA-SE I 2017 Immune Design
NCT00623831 Mixed bacteria vaccine I 2017 Ludwig Institute for Cancer Research
NCT00373217 MAGE-A1, Her-2/neu, FBP peptides ovarian cancer vaccine II 2016 Craig L Slingluff, Jr
National Cancer Institute (NCI)
NCT01522820 DEC-205/NY-ESO-1 Fusion Protein CDX-1401 I 2016 Roswell Park Cancer Institute
NCT01673217 NY-ESO-1 peptide vaccine I 2014 Roswell Park Cancer Institute
NCT00066729 NY-ESO-1 peptide vaccine I 2011 Memorial Sloan Kettering Cancer Center

Expression of classic CTA in ovarian cancer and research progress

The classical CTAs induce a robust immune response in several cancers. Their robust potential in the diagnosis, treatment, and outcome of ovarian cancer has garnered substantial attention. As a result, they have become the most widely studied classical CTAs.

MAGE-A

The MAGE-A subfamily of the MAGE gene family was the first classical protein identified as a CTA. It is encoded by genes located on the X chromosome (MAGE-A1 to MAGE-A12) [19,20]. Similar to other CTA family members, MAGE-A is rarely expressed in normal tissues but is highly expressed in bladder cancer [21,22], lung cancer [23], skin cancer [24], and other tumor tissues. These proteins show significantly high expression and strong immunogenicity in epithelial ovarian cancer (EOC) [20,25,26]. Schooten et al. reported that antigen peptides encoded by MAGE-A family genes are presented to T cells by MHC-I molecules of tumor cells through dendritic cells (DC) to activate effector T cells then exert specific anti-tumor activity [27]. These findings suggest that immunotherapy with MAGE-A is a promising therapy.

The expression of MAGE-A in ovarian cancer

Expression levels of MAGE in ovarian cancer have been studied. Daudi et al. used the reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry to measure the expression of MAGE-A in 400 EOC tissues and found that at least five MAGE-A family members (MAGE-A1, MAGE-A3, MAGE-A4, MAGE-A10, and MAGE-C1) are expressed in approximately 78% of EOC patients [28]. In particular, MAGE-A4 shows relatively high expression frequency and plays a central role in the co-expression of other MAGE-A antigens [29]. Another study showed that, compared with patients with benign diseases, serum levels of MAGE-A4 in ovarian cancer patients were significantly higher, and MAGE-A4 protein was expressed in nearly 22% of primary patients [30]. Yamada et al. reported that the frequency of MAGE-A1 mRNA expression in 58 ovarian cancer tissues was 20.7% [28]. Szajnik et al. reported that MAGE-A3/6 protein was present in all plasma-derived exosomes of ovarian cancer patients but not in benign tumors or healthy controls [31]. Hofmann et al. used multiplex RT-PCR analysis to measure BAGE, MAGE-A1, MAGE-A3, and GAGE1/2 mRNA in peritoneal fluid in patients with ovarian cancer. The combination of the four markers showed a 94% increase in diagnostic sensitivity for ovarian cancer compared to cell morphology alone [32]. Analysis of the Oncomine dataset revealed that MAGE-A is also co-expressed with other genes in EOC, including CT45 [33].

In summary, the expression of MAGE-A is tumor-specific. Compared with benign disease or healthy controls, MAGE-A is highly expressed in tissues and serum of patients. MAGE family members can be used as combined markers for early diagnosis. The high levels of MAGE-A expression in ovarian cancer allow researchers to investigate its value as an outcome predictor in ovarian cancer.

Correlation between MAGE-A and outcomes in ovarian cancer

Many studies reported significant correlations between CTAs and tumor progression and outcome, including for members of the MAGE-A family. Zhang et al. reported that the expression of MAGE-A1 and MAGE-A3 is related to the degree of tumor differentiation and clinical stage of ovarian cancer [34]. Daudi et al. found that, in EOC, the expression of MAGE-A1 and MAGE-A10 was significantly correlated with shorter progression-free survival [29]. Yakirevich et al. demonstrated that MAGE-A4 expression was present in 57% of high-grade serous ovarian cancer, and no staining was detected in serous cystadenoma or normal ovary. MAGE-A4 expression was negatively correlated with survival. Multivariate analysis showed that the expression of MAGE-A4 is an independent risk factor for outcomes; the authors believe that MAGE-A4 may be a reliable prognostic indicator for patients with serous ovarian cancer [35]. The expression of MAGE-A9 is closely related to the high histopathological grade, International Federation of Gynaecology and Obstetrics (FIGO) stage, CA-125 level, and metastasis of ovarian cancer. Patients with MAGE-A9 expression showed poor overall survival [36]. A study examined the prognostic significance of MAGE-A expression in patients with EOC and found that MAGE-A expression is related to the pathological type, FIGO stage, and preoperative serum CA125 levels. Compared with patients with EOC that are MAGE-A-negative, the overall survival of EOC with expression of the MAGE-A family was significantly shorter, consistent with previous findings [20]. Coincidentally, Sang et al. showed that the expression levels of each MAGE-A gene in the peripheral blood of ovarian cancer patients are associated with lower overall survival [26]. Consistently, Daudi et al. used enzyme-linked immunosorbent assay for MAGE-A antigen-specific antibody and found that the presence of humoral immune response against any MAGE antigen signals poor outcome [29].

These studies suggest that MAGE-A is highly expressed in EOC and affects the occurrence, progression, and outcome of epithelial ovarian cancer; for these reasons, MAGE-A has the potential to be a specific target for immunotherapy against ovarian cancer.

Research into MAGE-A in immunotherapy of ovarian cancer

Therapeutic tumor vaccines may be used to exploit CTAs to inhibit or delay tumor growth. After years of research, many therapeutic tumor vaccines have been developed and used, including protein or peptide vaccines, cell-based vaccines, DNA or RNA vaccines, and vector-based vaccines [27]. The MAGE-A protein family has attracted extensive attention in exploring CTA-based immunotherapy because of its expression characteristics. These antigens belong to a highly conserved proteome and share a common MAGE domain that is widely expressed in several tumors [37,38]. A pre-clinical study showed that, among 15 genetically diverse and unrelated mice immunized with the MAGE-A vaccine, 14 were induced to respond and develop a cross-reactive immune response. The MAGE-A DNA therapeutic vaccine significantly slowed the growth of tumors and doubled the median survival rate in mice. These findings support the clinical application of several MAGE-A family members to avoid tumor immune escape [39]. Because DCs are the primary antigen-presenting cells, and antigens presented by them activate T cells, many studies focused on peptide-incubated DC vaccines [40]. This therapeutic strategy has been used in clinical trials of the MAGE-A vaccine in melanoma [41-46] and some clinical trials of lung cancer [46-49], colon cancer [50], and myeloma [51]. However, there are few reports on clinical trials of targeted MAGE-A vaccines directly against ovarian cancer. As one of the most immunogenic MAGE-A proteins, the CTA vaccine targeted by MAGE-A4 is being evaluated in a clinical trial, and it targets a broad spectrum of cancers [52]. Batchu et al. used DCs transduced with rAAV-6 capsid mutation vector Y445F to induce MAGE-A3-specific anti-tumor cytotoxic T lymphocyte responses in vitro. This form of rAAV-based DC immunotherapy targeting MAGE-A3, whether used alone or in combination with other immune enhancement programs, may prove effective in treating EOC [53].

The DNA therapeutic vaccine and DC cell vaccine showed promising results. In recent years, clinical trials of MAGE-A targeted tumor immunotherapy has been widely carried out. There are ongoing clinical safety evaluations of T cell therapy in HLA-A2+ subjects with high MAGE-A4 expression and efficacy evaluations experiment of MAGE-A4 T cell therapy combined with low-dose radiation, including in patients with ovarian cancer [54]. In the future, MAGE-A will provide promising new opportunities to treat ovarian cancer.

NY-ESO-1

NY-ESO-1, also known as cancer-testis antigen 1B, is one of the members of the CTA gene family and is encoded by a gene from the Xq28 chromosome located in the coding region of other CTA (MAGE family members) [55,56]. Chen et al. first described the significance of NY-ESO-1 as a tumor-associated antigen. In 1997, the authors used the cDNA of a patient with esophageal squamous cell carcinoma as an antigen in the patient’s serum, then measured the humoral response and identified the corresponding antibody. They also measured the humoral reaction of the antigen in various tumors and found positive reactions in melanoma, ovarian cancer, breast cancer, and bladder cancer [10,57]. Similar to MAGE-A4 [58], NY-ESO-1 can be identified in testicular germ cells at 18 weeks of gestation. In developing spermatogonia, NY-ESO-1 was identified as one of the nine genes upregulated during differentiation, suggesting its role in the clonal proliferation of spermatogonia [59].

The expression of NY-ESO-1 in ovarian cancer

The characteristics of NY-ESO-1 are like those of other CTAs. It is not expressed in normal tissues but is selectively expressed in testis and many malignant tumors [55], including melanoma [60,61], lung cancer [62,63], esophageal cancer [10], and ovarian cancer [64]. Odunsi et al. reported that about 50% of 107 EOC tissue samples expressed NY-ESO-1 or LAGE-1. NY-ESO-1 and LAGE-1 were co-expressed in 11% epithelial ovarian tumor tissue samples and SKOV3 cells [64]. Subsequent studies reported CTA analyses in many ovarian cancer patients and found that NY-ESO-1 was expressed in 41% of tumors [65]. Hurley et al. found that the combination of TRIM21 and NY-ESO-1 provided 67% sensitivity and 94% specificity; that is, TRIM21 and NY-ESO-1 can complement one another for screening women with genetic risk for ovarian cancer or for achieving an early diagnosis of ovarian cancer [66]. These results suggest that NY-ESO-1 is expressed in ovarian cancer patients and lays a foundation for studying the correlation between NY-ESO-1 expression and the occurrence, progression, and outcomes in ovarian cancer.

The correlation between NY-ESO-1 and outcomes in ovarian cancer

Szender et al. were the first to demonstrate the association between NY-ESO-1 expression and the malignant phenotype of invasive ovarian cancer. They reported that patients with positive NY-ESO-1 expression had more serous ovarian cancer tissue types and histopathological grade three tumor patients and higher stage and were less likely to achieve good responses to standard treatment. In patients with positive NY-ESO-1 expression, progression-free survival tended to be shorter, while OS was significantly shortened [65]. Although these data show that the expression of NY-ESO-1 is related to malignant degree, the specifically related factors still have substantial research value: for example, it is unknown whether there is a correlation between the high expression of NY-ESO-1 and the age and gender of patients with ovarian cancer; it is also unknown whether there is co-expression of NY-ESO-1 with confirmed indicators of poor outcome; finally, it is unclear whether there is expression of NY-ESO-1 in patients with ovarian cancer before and after traditional treatment (surgery, radiotherapy, and chemotherapy).

Research progress of NY-ESO-1 in immunotherapy of ovarian cancer

Clinical trials of treatment strategies targeting NY-ESO-1 have been carried out in patients with ovarian cancer, and NY-ESO-1-specific immunotherapy has evident clinical benefits [67]. As expected, the NY-ESO-1-specific therapeutic vaccine-induced specific cellular and humoral immune responses in most NY-ESO-1-positive patients. Odunsi et al. reported that NY-ESO-1 with HLA-I and II dual-specificity can induce immune responses of complete antibody, HLA-DP4 restricted CD4+ T cells, and HLA-A2/A24 restricted CD8+ T cells in patients with ovarian cancer [68]. After seven years of clinical trials, these researchers reported encouraging results. The combined treatment strategy of decitabine and NY-ESO-1 vaccine stabilized the disease in patients with recurrent EOC or achieved partial clinical responses [69]. Decitabine may enhance the immune response to the NY-ESO-1 vaccine by promoting demethylation of the CTA promoter and enhancing immunity [70,71]. The success of this regimen may supplement the standard second-line treatment strategy for EOC. Based on the mechanism of demethylation enhancing vaccine effect, Griffiths et al. reported that SGI-110 treatment-induced hypomethylation and CTA gene expression in a dose-dependent manner in vivo and in vitro was generally better than azacytidine or decitabine. There was enhanced expression of MHC-I and ICAM-1 and enhanced recognition of EOC cells by NY-ESO-1-specific CD8+ T cells [72]. These findings suggest that SGI-110 is a candidate for combination with the NY-ESO-1-specific vaccine in EOC treatment.

NY-ESO-1 is an excellent tumor-associated antigen for immunotherapy (cancer vaccine) in clinical trials of immunotherapy for ovarian cancer. In addition to immunotherapy targeting NY-ESO-1, there is synergistic anti-cancer efficacy of NY-ESO-1 with known anti-cancer medications, and this has been confirmed in several medical institutions. There are promising results of NY-ESO-1 in phase I/II clinical trials, especially in ovarian cancer.

CT45

The cancer-testis antigen-45 family (CT45) is not expressed in normal tissues (except for the testis) but is abnormally expressed in many cancers [11]. Large-scale parallel signature sequencing and RT-PCR methods were used for screening and evaluation, and six genes with high similarity (>98% cDNA consistency) were obtained, including the CT45 family (CT45A1, CT45A2, CT45A3, CT45A4, CT45A5, and CT45A6) that is clustered in series in the 125 kb region of xq26.3 [73]. There are many reports of CT45A1. For example, when breast cancer was used as a research model, it was found that the overexpression of CT45A1 positively correlated with tumor invasion and metastasis [11]. Yang et al. reported that MAGE-D4B, CAGE, and CT45A1 promoted epithelial-mesenchymal transition (EMT) and metastasis via upregulation of EMT and metastatic genes [74]. These findings suggest that CT45 has a high research value as a potential cancer target.

The expression of CT45 in ovarian cancer

Chen et al. found that the expression of CT45 protein in tumors correlated with mRNA level detected by quantitative RT-PCR. The evaluation of CT45 protein expression on a variety of cancer tissue microarrays (376 cases of lung cancer, 219 cases of ovarian cancer, and 155 cases of breast cancer) showed that CT45 was expressed most frequently in ovarian cancer (37%), followed by lung cancer (13%), the lowest in breast cancer (<5%) [75]. Zhang et al. identified the co-expression of CT45 and other CTAs through analysis of the Oncomine data set; these genes include MAGE and GAGE [33].

The correlation between CT45 and outcomes in ovarian cancer

Koop et al. studied the biological functions of CT45 cells and found that the increased activity of CT45-positive cells was significantly associated with the progression of the disease [76]. Although the expression mechanism of CT45 in cancer has not yet been elucidated, the relationship between the expression of CT45 and the development of ovarian cancer and survival has been studied by DNA methylation. After Zhang et al. determined the expression of CT45 in EOC, they defined the epigenetic regulation of CT45 through DNA methylation. Subsequent studies confirmed that, in EOC, CT45 promoter hypomethylation was associated with reduced OS, suggesting that CT45 expression and promoter hypomethylation may indicate poor outcomes in patients with ovarian cancer [33]. Coscia et al. used mass spectrometry-based proteomics and found that CT45 can be used as a marker for long-term survival in advanced metastatic advanced serous ovarian cancer after chemotherapy [77]. There remains controversy regarding the correlation between CT45 and outcomes in ovarian cancer, possibly related to disease state and other treatments.

Research on CT45 in immunotherapy of ovarian cancer

To improve long-term survival and outcomes in immunogenic tumors such as EOC, various anti-tumor vaccines are being developed. Chen et al. showed that the frequency and characteristics of CT45 expression are similar to other CTA cancer vaccine targets in clinical trials (i.e., NY-ESO-1 and MAGE-A) [75]. Other studies showed that decitabine treatment induces the expression of CT45 mRNA and protein in EOC cells, and promoter transgenic analysis showed that DNA methylation directly inhibits the activity of the CT45 promoter. These results suggest that decitabine or other epigenetic regulators might provide effective immune targeting for CT45 [33]. Coscia et al. found that CT45-derived HLA-I peptides using immunopeptideomics activated patient-derived cytotoxic T cells and promoted tumor cell killing [77].

Thus far, our summary suggests that cancer vaccines alone and in combination with anti-tumor medications can improve anti-tumor efficacy. Clinical research on NY-ESO-1 is currently relatively extensive. We can use this as a reference to expand the study of CT45 antigen as a tumor immunotherapy target and explore prospects for its clinical application.

Sp17

Sp17 is a highly conserved protein composed of 151 amino acids. It was initially isolated from rabbit epididymal sperm and testicular membrane sediments, and it has high interspecies homology [78-80]. It was initially thought that Sp17 was only expressed in the testis, and its primary function was to bind to the extracellular matrix of oocytes, allowing sperm to interact with the zona pellucida [81]. Some reports suggested that Sp17 participates in heparin sulfate-mediated cell adhesion or migration in transformed lymphocytes and heme cells [82]. Sp17 is highly expressed in ovarian cancer [83], head and neck squamous cell carcinoma [84], breast cancer [17], non-small cell lung cancer [85], multiple myeloma [86,87], and other hematological malignancies [88]. These findings suggest that Sp17 may be a highly immunogenic autoantigen in humans and can be used as a target for anti-tumor immunotherapy.

The expression of Sp17 in ovarian cancer

Sp17 was first found to be highly specifically expressed in multiple myeloma. As research extended, Sp17 expression characteristics in ovarian cancer became more apparent [87]. Straughn et al. detected Sp17 transcripts in 15 of 18 cases of primary ovarian cancer (83%), while Sp17 transcripts were not detected in normal cervix or cervix. Sp17 protein was detected in eight of 19 normal sperm and 19 ovarian cancer tissue samples, primarily located in the nucleus with a small amount of cytoplasmic staining [89]. Li et al. showed that Sp17 was abnormally expressed in 43% (30/70) of patients with primary EOC and eight patients with cancer cell-containing ascites. Sp17 expression was also detected in metastatic lesions. In addition, the authors found that overexpression of HSp17 increased the ability of ovarian cancer cells to migrate [90]. These studies suggest that the expression of Sp17 in EOC tissues is increased and suggest that Sp17 is a potential biomarker of EOC.

The correlation between Sp17 and outcomes in ovarian cancer

Brunette et al. showed that Sp17 is not significantly related to the staging of ovarian cancer, it has a higher expression in benign and borderline serous tumors, and its expression further decreases as the cancer level increases [83]. The authors also analyzed the failure free survival and overall survival of 336 EOC patients and found no significant correlation between Sp17 expression and failure free survival or overall survival [83]. However, small studies found that high expression of Sp17 reduced the chemosensitivity of EOC cells to carboplatin and cisplatin [90]. Sp17 is defined as a candidate gene related to chemotherapy resistance for clear cell adenocarcinoma. Downregulation of Sp17 significantly increased the sensitivity of ovarian cancer cells to paclitaxel [91,92]. Although the overall outcome of borderline serous and low-grade malignant tumors is better than that of high-grade malignant tumors, they are also chemotherapy-resistant tumors, and it is difficult to treat them if they are not treated with complete surgical resection. Therefore, if there is an effective targeted therapy for Sp17, patients with positive expression of this protein may experience improved outcomes.

Sp17 in immunotherapy of ovarian cancer

Considering the characteristics and expression limitations of Sp17 protein itself, strategies for anti-ovarian cancer immunotherapy against Sp17 are also emerging. Gao et al. found that, compared with Sp17low (PD-L1-MHCII+) EOC cells, the Sp17high (PD-L1+MHCII-) EOC cell population showed significantly enhanced resistance to paclitaxel-induced cell death in vitro. This finding suggests that Sp17 can become a target of immunotherapy [92]. Chiriva-Internati et al. successfully produced HLA-I restricted CTLs from the peripheral blood of three patients with ovarian cancer that target and recognize Sp17. This specific CTL can cleave Sp17 positive autologous tumor cells by modulating the perforin pathway and secreting INF-γ and a small amount of IL-4 [12]. Sp17-conjugated CpG as a therapeutic and preventive vaccine can activate T cells in vivo and redirect inhibitory T-regs to the activated Th-17 phenotype. This vaccine strategy may achieve tumor control and increased survival in ovarian cancer patients [93]. Researchers constructed an antibody-drug conjugate (Sp17-doxorubicin) and found that it inhibited the activity of cancer cells and induced the regression of established SKOV-3 xenograft tumors in mice [94].

In the study of Sp17 immunotherapy, the application of tumor antigen epitope peptides received the most attention. Vermeij et al. used tissue microarrays to determine MHC class I and tumor antigens (including p53, Sp17, survivin, WT1, and NY-ESO-1) in 270 primary tumors tissue samples. In 74.3% of these primary tumors, the co-expression of MHC-I and at least one tumor antigen was observed. These results provide a research basis for multi-epitope anti-ovarian cancer immunotherapy [95].

In recent years, there have also been reports on methods for obtaining potential epitope peptides through big data analysis and screening with software such as the Pir Peptide Match program [96]. Xiang et al. confirmed the immunogenicity of six epitope peptides from Sp17 under the mode of epitope peptide-conjugated CpG and found that the epitope peptide from the 111-142aa sequence of hSp17 induced high levels of antibodies and T cells expressing IFN-γ. One tumor-bearing transgenic mouse treated with the corresponding drug experienced a significantly prolonged lifespan [97]. Mattila et al. optimized the dosage form and incorporated Sp17-derived antigen peptides and CpG oligonucleotides into spray-dried microparticles, administered in oral form. Analysis of spleen cells harvested from vaccinated tumor-bearing mice showed that the immune system responds to SP17 antigen re-stimulation, IFN-γ+/CD8+ lymphocytes show strong activation. Four weeks after the tumor challenge, the administration group showed significant ascites/tumor volume reduction [98].

These studies demonstrate that immunotherapy based on Sp17 antigen focus on increasing immunogenicity, enhancing broad-spectrum, and increasing effectiveness. We focused on practical problems encountered in clinical application to optimize preparation and administration modes. Although no anti-ovarian cancer drug targeting Sp17 has entered the clinical trial stage, it can be expected that Sp17 will soon emerge as a target with excellent expression and immunological properties.

The relationships among these CTAs

Based on these four classic CTAs expressed in ovarian cancer, we conclude that expression of the four classic CTAs differ in expression patterns between patients with cancer and normal controls. Second, CTAs can be divided into X-CTA and non-X-CTA depending on whether CTA is located on the X chromosome. The four CTAs referenced above are all located on the X chromosome. It is unknown whether different CTA localizations play different roles in ovarian cancer or predict different outcomes; however, the question is worthy of further study. Next, the four CTAs exist as multiple family members, and analysis shows that the frequency of expression differs among family members. Finally, the four CTAs induce cellular and humoral immune responses. Taken together, the evidence suggests that these CTAs can be used as tumor markers in ovarian cancer and may serve as targets for cancer vaccines (Table 2).

Table 2.

The relationships between these CTAs

Species Expression pattern Positioning Composition Immune correlation
MAGE-A It is not expressed in normal tissues, but only in normal testis and a variety of tumor tissues X chromosome Often in the form of multiple family members Cause cellular and humoral immune response
NY-ESO-1
CT45
Sp17

Relationship between newly discovered CTAs and ovarian cancer

With the development of cancer vaccine research, several new tumor antigens have been identified, including several kinds of CTA. Xu et al. found that knocking down TEX19 inhibited the proliferation, migration, and invasion of ovarian cancer cells. A peptide derived from the dominant epitope of TEX19 may serve as a target for an anti-tumor vaccine [13]. Gupta et al. showed that HSP70-2 promotes the proliferation, migration, and invasion of ovarian cancer cells. For the first time, it was found that ovarian cancer cells lacking HSP70-2 reduced cell motility due to low expression of EMT molecules [16].

The POTE family genes are divided into three phylogenetic groups, Group I (POTEA), Group II (POTEB1, B2, B3, C, and D), and Group III (POTE, F, I, J, KP, and M) [15]. Barger et al. found that POTE family genes (especially group III POTE members) are highly expressed in ovarian cancer tissues and are related to poor outcomes [14]. As new CTAs are identified as potential targets and biomarkers for cancer treatment, the means and methods for cancer treatment will be enriched. Regardless of whether it is used as a single-gene tumor antigen for cancer vaccines or as a combination therapy with other cancer treatments, CTA plays an essential role in treating ovarian cancer.

Discussion

Because ovarian cancer is usually diagnosed at advanced stages and has relatively high postoperative recurrence and metastasis rates, it is difficult to achieve satisfactory results using conventional therapy alone. In ovarian cancer, control of tumor cell growth and removal of residual tumor cells after surgery or chemotherapy remain critical subjects of future study. We urgently need effective treatment strategies (particularly tumor immunotherapy) to resolve this problem. Clinical trials have shown that tumor immunotherapy is effective for ovarian cancer, suggesting a direction for developing ovarian cancer treatment strategies in the future.

Although tumor immunotherapy is an excellent modality for treating ovarian cancer, there remain some problems for individual patients. MAGE-A, NY-ESO-1, CT45, and Sp17 are not expressed in all ovarian cancers. Furthermore, it remains unknown whether the expression of these antigens in ovarian cancer patients can trigger effective immune responses to eliminate tumor cells; it is also unknown whether there are more effective epitope peptides of MAGE-A NY-ESO-1, CT45, and Sp17 that can stimulate sufficient immune responses. To resolve these problems, we should continue to screen and develop other members of the CTA family with high expression in ovarian cancer to cover more ovarian cancer patients. We should develop highly efficient and low-toxicity epigenetic modifiers to enhance the immune effect of CTA vaccines. Improved use of big data and bioinformatics technology will result in the design of optimal candidate epitopes of MAGE-A, NY-ESO-1, CT45, Sp17, and other CTAs to significantly enhance the tumor-killing ability of CTL cells.

The role of cancer/testicular antigens in the development and etiology of ovarian cancer is not yet clear. An in-depth study and explanation of the biological functions of these CTAs and the interaction between their mechanisms will lay a solid foundation for ovarian cancer immunotherapy with CTA as the target. At present, many research institutions are engaged in this research. Although the results are not completely satisfactory, we believe that immunotherapy will become an essential strategy for treating ovarian cancer in the future.

Acknowledgements

The review was supported by NSFC joint fund for regional innovation and development [No. U20A20413]; Natural Science Fund of Liaoning Province of China (No. 2020-M S-141); Scientific Research Fund of Liaoning Provincial Education Department of China (No. QN2019033); Major Special S&T Projects in Liaoning Province (2019JH1/10300005); Shenyang S&T Projects (20-204-4-22); Program for Shenyang High Level Talent Innovation and Entrepreneurship Team (2019-SYRCCY-B-01); Shenyang S&T Projects (19-109-4-09). The authors thank the Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Cancer of Ministry of Education (China Medical University) for its support.

Disclosure of conflict of interest

None.

References

  • 1.La Vecchia C. Ovarian cancer: epidemiology and risk factors. Eur J Cancer Prev. 2017;26:55–62. doi: 10.1097/CEJ.0000000000000217. [DOI] [PubMed] [Google Scholar]
  • 2.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7–30. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
  • 3.Bell JG. Early detection of ovarian cancer: if only we had a “Pap smear” for this disease. Cancer. 2011;117:4340–4342. doi: 10.1002/cncr.26025. [DOI] [PubMed] [Google Scholar]
  • 4.Xie K, Fu C, Wang S, Xu H, Liu S, Shao Y, Gong Z, Wu X, Xu B, Han J, Xu J, Xu P, Jia X, Wu J. Cancer-testis antigens in ovarian cancer: implication for biomarkers and therapeutic targets. J Ovarian Res. 2019;12:1. doi: 10.1186/s13048-018-0475-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Coukos G, Tanyi J, Kandalaft LE. Opportunities in immunotherapy of ovarian cancer. Ann Oncol. 2016;27(Suppl 1):i11–i15. doi: 10.1093/annonc/mdw084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zhang Y, Zhang Y, Zhang L. Expression of cancer-testis antigens in esophageal cancer and their progress in immunotherapy. J Cancer Res Clin Oncol. 2019;145:281–291. doi: 10.1007/s00432-019-02840-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Whitehurst AW. Cause and consequence of cancer/testis antigen activation in cancer. Annu Rev Pharmacol Toxicol. 2014;54:251–272. doi: 10.1146/annurev-pharmtox-011112-140326. [DOI] [PubMed] [Google Scholar]
  • 8.Gordeeva O. Cancer-testis antigens: unique cancer stem cell biomarkers and targets for cancer therapy. Semin Cancer Biol. 2018;53:75–89. doi: 10.1016/j.semcancer.2018.08.006. [DOI] [PubMed] [Google Scholar]
  • 9.Wei X, Chen F, Xin K, Wang Q, Yu L, Liu B, Liu Q. Cancer-testis antigen peptide vaccine for cancer immunotherapy: progress and prospects. Transl Oncol. 2019;12:733–738. doi: 10.1016/j.tranon.2019.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Oshima Y, Shimada H, Yajima S, Nanami T, Matsushita K, Nomura F, Kainuma O, Takiguchi N, Soda H, Ueda T, Iizasa T, Yamamoto N, Yamamoto H, Nagata M, Yokoi S, Tagawa M, Ohtsuka S, Kuwajima A, Murakami A, Kaneko H. NY-ESO-1 autoantibody as a tumor-specific biomarker for esophageal cancer: screening in 1969 patients with various cancers. J Gastroenterol. 2016;51:30–34. doi: 10.1007/s00535-015-1078-8. [DOI] [PubMed] [Google Scholar]
  • 11.Shang B, Gao A, Pan Y, Zhang G, Tu J, Zhou Y, Yang P, Cao Z, Wei Q, Ding Y, Zhang J, Zhao Y, Zhou Q. CT45A1 acts as a new proto-oncogene to trigger tumorigenesis and cancer metastasis. Cell Death Dis. 2014;5:e1285. doi: 10.1038/cddis.2014.244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chiriva-Internati M, Wang Z, Salati E, Timmins P, Lim SH. Tumor vaccine for ovarian carcinoma targeting sperm protein 17. Cancer. 2002;94:2447–2453. doi: 10.1002/cncr.10506. [DOI] [PubMed] [Google Scholar]
  • 13.Xu Z, Tang H, Zhang T, Sun M, Han Q, Xu J, Wei M, Yu Z. TEX19 promotes ovarian carcinoma progression and is a potential target for epitope vaccine immunotherapy. Life Sci. 2020;241:117171. doi: 10.1016/j.lfs.2019.117171. [DOI] [PubMed] [Google Scholar]
  • 14.Qazi S, Raza K. Phytochemicals from Ayurvedic plants as potential medicaments for ovarian cancer: an in silico analysis. J Mol Model. 2021;27:114. doi: 10.1007/s00894-021-04736-x. [DOI] [PubMed] [Google Scholar]
  • 15.Barger CJ, Zhang W, Sharma A, Chee L, James SR, Kufel CN, Miller A, Meza J, Drapkin R, Odunsi K, Klinkebiel D, Karpf AR. Expression of the POTE gene family in human ovarian cancer. Sci Rep. 2018;8:17136. doi: 10.1038/s41598-018-35567-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gupta N, Jagadish N, Surolia A, Suri A. Erratum: heat shock protein 70-2 (HSP70-2) a novel cancer testis antigen that promotes growth of ovarian cancer. Am J Cancer Res. 2020;10:1919–1920. [PMC free article] [PubMed] [Google Scholar]
  • 17.Mirandola L, Pedretti E, Figueroa JA, Chiaramonte R, Colombo M, Chapman C, Grizzi F, Patrinicola F, Kast WM, Nguyen DD, Rahman RL, Daver N, Ruvolo P, Post SM, Bresalier RS, Chiriva-Internati M. Cancer testis antigen Sperm Protein 17 as a new target for triple negative breast cancer immunotherapy. Oncotarget. 2017;8:74378–74390. doi: 10.18632/oncotarget.20102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kochin V, Kanaseki T, Tokita S, Miyamoto S, Shionoya Y, Kikuchi Y, Morooka D, Hirohashi Y, Tsukahara T, Watanabe K, Toji S, Kokai Y, Sato N, Torigoe T. HLA-A24 ligandome analysis of colon and lung cancer cells identifies a novel cancer-testis antigen and a neoantigen that elicits specific and strong CTL responses. Oncoimmunology. 2017;6:e1293214. doi: 10.1080/2162402X.2017.1293214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Meek DW, Marcar L. MAGE-A antigens as targets in tumour therapy. Cancer Lett. 2012;324:126–132. doi: 10.1016/j.canlet.2012.05.011. [DOI] [PubMed] [Google Scholar]
  • 20.Sang M, Wu X, Fan X, Lian Y, Sang M. MAGE-A family serves as poor prognostic markers and potential therapeutic targets for epithelial ovarian cancer patients: a retrospective clinical study. Gynecol Endocrinol. 2017;33:480–484. doi: 10.1080/09513590.2017.1291611. [DOI] [PubMed] [Google Scholar]
  • 21.Mengus C, Schultz-Thater E, Coulot J, Kastelan Z, Goluza E, Coric M, Spagnoli GC, Hudolin T. MAGE-A10 cancer/testis antigen is highly expressed in high-grade non-muscle-invasive bladder carcinomas. Int J Cancer. 2013;132:2459–2463. doi: 10.1002/ijc.27914. [DOI] [PubMed] [Google Scholar]
  • 22.Picard V, Bergeron A, Larue H, Fradet Y. MAGE-A9 mRNA and protein expression in bladder cancer. Int J Cancer. 2007;120:2170–2177. doi: 10.1002/ijc.22282. [DOI] [PubMed] [Google Scholar]
  • 23.Ayyoub M, Memeo L, Alvarez-Fernandez E, Colarossi C, Costanzo R, Aiello E, Martinetti D, Valmori D. Assessment of MAGE-A expression in resected non-small cell lung cancer in relation to clinicopathologic features and mutational status of EGFR and KRAS. Cancer Immunol Res. 2014;2:943–948. doi: 10.1158/2326-6066.CIR-13-0211. [DOI] [PubMed] [Google Scholar]
  • 24.Schultz-Thater E, Piscuoglio S, Iezzi G, Le Magnen C, Zajac P, Carafa V, Terracciano L, Tornillo L, Spagnoli GC. MAGE-A10 is a nuclear protein frequently expressed in high percentages of tumor cells in lung, skin and urothelial malignancies. Int J Cancer. 2011;129:1137–1148. doi: 10.1002/ijc.25777. [DOI] [PubMed] [Google Scholar]
  • 25.Kerkar SP, Wang ZF, Lasota J, Park T, Patel K, Groh E, Rosenberg SA, Miettinen MM. MAGE-A is more highly expressed than NY-ESO-1 in a systematic immunohistochemical analysis of 3668 cases. J Immunother. 2016;39:181–187. doi: 10.1097/CJI.0000000000000119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sang M, Wu X, Fan X, Sang M, Zhou X, Zhou N. Multiple MAGE-A genes as surveillance marker for the detection of circulating tumor cells in patients with ovarian cancer. Biomarkers. 2014;19:34–42. doi: 10.3109/1354750X.2013.865275. [DOI] [PubMed] [Google Scholar]
  • 27.Schooten E, Di Maggio A, van Bergen En Henegouwen PMP, Kijanka MM. MAGE-A antigens as targets for cancer immunotherapy. Cancer Treat Rev. 2018;67:54–62. doi: 10.1016/j.ctrv.2018.04.009. [DOI] [PubMed] [Google Scholar]
  • 28.Yamada A, Kataoka A, Shichijo S, Kamura T, Imai Y, Nishida T, Itoh K. Expression of MAGE-1, MAGE-2, MAGE-3/-6 and MAGE-4a/-4b genes in ovarian tumors. Int J Cancer. 1995;64:388–393. doi: 10.1002/ijc.2910640607. [DOI] [PubMed] [Google Scholar]
  • 29.Daudi S, Eng KH, Mhawech-Fauceglia P, Morrison C, Miliotto A, Beck A, Matsuzaki J, Tsuji T, Groman A, Gnjatic S, Spagnoli G, Lele S, Odunsi K. Expression and immune responses to MAGE antigens predict survival in epithelial ovarian cancer. PLoS One. 2014;9:e104099. doi: 10.1371/journal.pone.0104099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kawagoe H, Yamada A, Matsumoto H, Ito M, Ushijima K, Nishida T, Yakushiji M, Itoh K. Serum MAGE-4 protein in ovarian cancer patients. Gynecol Oncol. 2000;76:336–339. doi: 10.1006/gyno.1999.5701. [DOI] [PubMed] [Google Scholar]
  • 31.Szajnik M, Derbis M, Lach M, Patalas P, Michalak M, Drzewiecka H, Szpurek D, Nowakowski A, Spaczynski M, Baranowski W, Whiteside TL. Exosomes in plasma of patients with ovarian carcinoma: potential biomarkers of tumor progression and response to therapy. Gynecol Obstet (Sunnyvale) 2013;(Suppl 4):3. doi: 10.4172/2161-0932.S4-003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hofmann M, Ruschenburg I. mRNA detection of tumor-rejection genes BAGE, GAGE, and MAGE in peritoneal fluid from patients with ovarian carcinoma as a potential diagnostic tool. Cancer. 2002;96:187–193. doi: 10.1002/cncr.10622. [DOI] [PubMed] [Google Scholar]
  • 33.Zhang W, Barger CJ, Link PA, Mhawech-Fauceglia P, Miller A, Akers SN, Odunsi K, Karpf AR. DNA hypomethylation-mediated activation of Cancer/Testis Antigen 45 (CT45) genes is associated with disease progression and reduced survival in epithelial ovarian cancer. Epigenetics. 2015;10:736–748. doi: 10.1080/15592294.2015.1062206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zhang S, Zhou X, Yu H, Yu Y. Expression of tumor-specific antigen MAGE, GAGE and BAGE in ovarian cancer tissues and cell lines. BMC Cancer. 2010;10:163. doi: 10.1186/1471-2407-10-163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yakirevich E, Sabo E, Lavie O, Mazareb S, Spagnoli GC, Resnick MB. Expression of the MAGE-A4 and NY-ESO-1 cancer-testis antigens in serous ovarian neoplasms. Clin Cancer Res. 2003;9:6453–6460. [PubMed] [Google Scholar]
  • 36.Xu Y, Wang C, Zhang Y, Jia L, Huang J. Overexpression of MAGE-A9 is predictive of poor prognosis in epithelial ovarian cancer. Sci Rep. 2015;5:12104. doi: 10.1038/srep12104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Weon JL, Potts PR. The MAGE protein family and cancer. Curr Opin Cell Biol. 2015;37:1–8. doi: 10.1016/j.ceb.2015.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Coulie PG, Karanikas V, Lurquin C, Colau D, Connerotte T, Hanagiri T, Van Pel A, Lucas S, Godelaine D, Lonchay C, Marchand M, Van Baren N, Boon T. Cytolytic T-cell responses of cancer patients vaccinated with a MAGE antigen. Immunol Rev. 2002;188:33–42. doi: 10.1034/j.1600-065x.2002.18804.x. [DOI] [PubMed] [Google Scholar]
  • 39.Duperret EK, Liu S, Paik M, Trautz A, Stoltz R, Liu X, Ze K, Perales-Puchalt A, Reed C, Yan J, Xu X, Weiner DB. A designer cross-reactive DNA immunotherapeutic vaccine that targets multiple MAGE-A family members simultaneously for cancer therapy. Clin Cancer Res. 2018;24:6015–6027. doi: 10.1158/1078-0432.CCR-18-1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Schuler PJ, Harasymczuk M, Visus C, Deleo A, Trivedi S, Lei Y, Argiris A, Gooding W, Butterfield LH, Whiteside TL, Ferris RL. Phase I dendritic cell p53 peptide vaccine for head and neck cancer. Clin Cancer Res. 2014;20:2433–2444. doi: 10.1158/1078-0432.CCR-13-2617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.van Baren N, Bonnet MC, Dreno B, Khammari A, Dorval T, Piperno-Neumann S, Lienard D, Speiser D, Marchand M, Brichard VG, Escudier B, Negrier S, Dietrich PY, Maraninchi D, Osanto S, Meyer RG, Ritter G, Moingeon P, Tartaglia J, van der Bruggen P, Coulie PG, Boon T. Tumoral and immunologic response after vaccination of melanoma patients with an ALVAC virus encoding MAGE antigens recognized by T cells. J. Clin. Oncol. 2005;23:9008–9021. doi: 10.1200/JCO.2005.08.375. [DOI] [PubMed] [Google Scholar]
  • 42.Bonehill A, Van Nuffel AM, Corthals J, Tuyaerts S, Heirman C, Francois V, Colau D, van der Bruggen P, Neyns B, Thielemans K. Single-step antigen loading and activation of dendritic cells by mRNA electroporation for the purpose of therapeutic vaccination in melanoma patients. Clin Cancer Res. 2009;15:3366–3375. doi: 10.1158/1078-0432.CCR-08-2982. [DOI] [PubMed] [Google Scholar]
  • 43.Wilgenhof S, Van Nuffel AM, Corthals J, Heirman C, Tuyaerts S, Benteyn D, De Coninck A, Van Riet I, Verfaillie G, Vandeloo J, Bonehill A, Thielemans K, Neyns B. Therapeutic vaccination with an autologous mRNA electroporated dendritic cell vaccine in patients with advanced melanoma. J Immunother. 2011;34:448–456. doi: 10.1097/CJI.0b013e31821dcb31. [DOI] [PubMed] [Google Scholar]
  • 44.Kruit WH, Suciu S, Dreno B, Mortier L, Robert C, Chiarion-Sileni V, Maio M, Testori A, Dorval T, Grob JJ, Becker JC, Spatz A, Eggermont AM, Louahed J, Lehmann FF, Brichard VG, Keilholz U. Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in metastatic melanoma. J. Clin. Oncol. 2013;31:2413–2420. doi: 10.1200/JCO.2012.43.7111. [DOI] [PubMed] [Google Scholar]
  • 45.Kranz LM, Diken M, Haas H, Kreiter S, Loquai C, Reuter KC, Meng M, Fritz D, Vascotto F, Hefesha H, Grunwitz C, Vormehr M, Husemann Y, Selmi A, Kuhn AN, Buck J, Derhovanessian E, Rae R, Attig S, Diekmann J, Jabulowsky RA, Heesch S, Hassel J, Langguth P, Grabbe S, Huber C, Tureci O, Sahin U. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534:396–401. doi: 10.1038/nature18300. [DOI] [PubMed] [Google Scholar]
  • 46.Grob JJ, Mortier L, D’Hondt L, Grange F, Baurain JF, Dreno B, Lebbe C, Robert C, Dompmartin A, Neyns B, Gillet M, Louahed J, Jarnjak S, Lehmann FF. Safety and immunogenicity of MAGE-A3 cancer immunotherapeutic with dacarbazine in patients with MAGE-A3-positive metastatic cutaneous melanoma: an open phase I/II study with a first assessment of a predictive gene signature. ESMO Open. 2017;2:e000203. doi: 10.1136/esmoopen-2017-000203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tyagi P, Mirakhur B. MAGRIT: the largest-ever phase III lung cancer trial aims to establish a novel tumor-specific approach to therapy. Clin Lung Cancer. 2009;10:371–374. doi: 10.3816/CLC.2009.n.052. [DOI] [PubMed] [Google Scholar]
  • 48.Vansteenkiste J, Zielinski M, Linder A, Dahabreh J, Gonzalez EE, Malinowski W, Lopez-Brea M, Vanakesa T, Jassem J, Kalofonos H, Perdeus J, Bonnet R, Basko J, Janilionis R, Passlick B, Treasure T, Gillet M, Lehmann FF, Brichard VG. Adjuvant MAGE-A3 immunotherapy in resected non-small-cell lung cancer: phase II randomized study results. J. Clin. Oncol. 2013;31:2396–2403. doi: 10.1200/JCO.2012.43.7103. [DOI] [PubMed] [Google Scholar]
  • 49.Vansteenkiste JF, Cho BC, Vanakesa T, De Pas T, Zielinski M, Kim MS, Jassem J, Yoshimura M, Dahabreh J, Nakayama H, Havel L, Kondo H, Mitsudomi T, Zarogoulidis K, Gladkov OA, Udud K, Tada H, Hoffman H, Bugge A, Taylor P, Gonzalez EE, Liao ML, He J, Pujol JL, Louahed J, Debois M, Brichard V, Debruyne C, Therasse P, Altorki N. Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2016;17:822–835. doi: 10.1016/S1470-2045(16)00099-1. [DOI] [PubMed] [Google Scholar]
  • 50.Takahashi N, Ohkuri T, Homma S, Ohtake J, Wakita D, Togashi Y, Kitamura H, Todo S, Nishimura T. First clinical trial of cancer vaccine therapy with artificially synthesized helper/ killer-hybrid epitope long peptide of MAGE-A4 cancer antigen. Cancer Sci. 2012;103:150–153. doi: 10.1111/j.1349-7006.2011.02106.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Xu YY, Kalos M, Cai L, Fang HB, Weiss BM, Badros A, Yanovich S, Akpek G, Tsao P, Cross A, Mann D, Philip S, Kerr N, Brennan A, Zheng Z, Ruehle K, Milliron T, Strome SE, Salazar AM, Levine BL, June CH. Combination immunotherapy after ASCT for multiple myeloma using MAGE-A3/Poly-ICLC immunizations followed by adoptive transfer of vaccine-primed and costimulated autologous T cells. Clin Cancer Res. 2014;20:1355–1365. doi: 10.1158/1078-0432.CCR-13-2817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Esfandiary A, Ghafouri-Fard S. MAGE-A3: an immunogenic target used in clinical practice. Immunotherapy. 2015;7:683–704. doi: 10.2217/imt.15.29. [DOI] [PubMed] [Google Scholar]
  • 53.Batchu RB, Gruzdyn OV, Moreno-Bost AM, Szmania S, Jayandharan G, Srivastava A, Kolli BK, Weaver DW, van Rhee F, Gruber SA. Efficient lysis of epithelial ovarian cancer cells by MAGE-A3-induced cytotoxic T lymphocytes using rAAV-6 capsid mutant vector. Vaccine. 2014;32:938–943. doi: 10.1016/j.vaccine.2013.12.049. [DOI] [PubMed] [Google Scholar]
  • 54.Sanderson JP, Crowley DJ, Wiedermann GE, Quinn LL, Crossland KL, Tunbridge HM, Cornforth TV, Barnes CS, Ahmed T, Howe K, Saini M, Abbott RJ, Anderson VE, Tavano B, Maroto M, Gerry AB. Preclinical evaluation of an affinity-enhanced MAGE-A4-specific T-cell receptor for adoptive T-cell therapy. Oncoimmunology. 2020;9:1682381. doi: 10.1080/2162402X.2019.1682381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jungbluth AA, Chen YT, Stockert E, Busam KJ, Kolb D, Iversen K, Coplan K, Williamson B, Altorki N, Old LJ. Immunohistochemical analysis of NY-ESO-1 antigen expression in normal and malignant human tissues. Int J Cancer. 2001;92:856–860. doi: 10.1002/ijc.1282. [DOI] [PubMed] [Google Scholar]
  • 56.Chen YT, Boyer AD, Viars CS, Tsang S, Old LJ, Arden KC. Genomic cloning and localization of CTAG, a gene encoding an autoimmunogenic cancer-testis antigen NY-ESO-1, to human chromosome Xq28. Cytogenet Cell Genet. 1997;79:237–240. doi: 10.1159/000134734. [DOI] [PubMed] [Google Scholar]
  • 57.Stockert E, Jager E, Chen YT, Scanlan MJ, Gout I, Karbach J, Arand M, Knuth A, Old LJ. A survey of the humoral immune response of cancer patients to a panel of human tumor antigens. J Exp Med. 1998;187:1349–1354. doi: 10.1084/jem.187.8.1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Satie AP, Rajpert-De Meyts E, Spagnoli GC, Henno S, Olivo L, Jacobsen GK, Rioux-Leclercq N, Jégou B, Samson M. The cancer-testis gene, NY-ESO-1, is expressed in normal fetal and adult testes and in spermatocytic seminomas and testicular carcinoma in situ. Lab Invest. 2002;82:775–780. doi: 10.1097/01.lab.0000017169.26718.5f. [DOI] [PubMed] [Google Scholar]
  • 59.von Kopylow K, Kirchhoff C, Jezek D, Schulze W, Feig C, Primig M, Steinkraus V, Spiess AN. Screening for biomarkers of spermatogonia within the human testis: a whole genome approach. Hum Reprod. 2010;25:1104–1112. doi: 10.1093/humrep/deq053. [DOI] [PubMed] [Google Scholar]
  • 60.Prasad ML, Jungbluth AA, Patel SG, Iversen K, Hoshaw-Woodard S, Busam KJ. Expression and significance of cancer testis antigens in primary mucosal melanoma of the head and neck. Head Neck. 2004;26:1053–1057. doi: 10.1002/hed.20112. [DOI] [PubMed] [Google Scholar]
  • 61.Aung PP, Liu YC, Ballester LY, Robbins PF, Rosenberg SA, Lee CC. Expression of New York esophageal squamous cell carcinoma-1 in primary and metastatic melanoma. Hum Pathol. 2014;45:259–267. doi: 10.1016/j.humpath.2013.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gure AO, Chua R, Williamson B, Gonen M, Ferrera CA, Gnjatic S, Ritter G, Simpson AJ, Chen YT, Old LJ, Altorki NK. Cancer-testis genes are coordinately expressed and are markers of poor outcome in non-small cell lung cancer. Clin Cancer Res. 2005;11:8055–8062. doi: 10.1158/1078-0432.CCR-05-1203. [DOI] [PubMed] [Google Scholar]
  • 63.Chen YT, Scanlan MJ, Sahin U, Tureci O, Gure AO, Tsang S, Williamson B, Stockert E, Pfreundschuh M, Old LJ. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A. 1997;94:1914–1918. doi: 10.1073/pnas.94.5.1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Odunsi K, Jungbluth AA, Stockert E, Qian F, Gnjatic S, Tammela J, Intengan M, Beck A, Keitz B, Santiago D, Williamson B, Scanlan MJ, Ritter G, Chen YT, Driscoll D, Sood A, Lele S, Old LJ. NY-ESO-1 and LAGE-1 cancer-testis antigens are potential targets for immunotherapy in epithelial ovarian cancer. Cancer Res. 2003;63:6076–6083. [PubMed] [Google Scholar]
  • 65.Szender JB, Papanicolau-Sengos A, Eng KH, Miliotto AJ, Lugade AA, Gnjatic S, Matsuzaki J, Morrison CD, Odunsi K. NY-ESO-1 expression predicts an aggressive phenotype of ovarian cancer. Gynecol Oncol. 2017;145:420–425. doi: 10.1016/j.ygyno.2017.03.509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hurley LC, Levin NK, Chatterjee M, Coles J, Muszkat S, Howarth Z, Dyson G, Tainsky MA. Evaluation of paraneoplastic antigens reveals TRIM21 autoantibodies as biomarker for early detection of ovarian cancer in combination with autoantibodies to NY-ESO-1 and TP53. Cancer Biomark. 2020;27:407–421. doi: 10.3233/CBM-190988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Weinberger V, Minár L. Immunotherapy in ovarian cancer. Ceska Gynekol. 2012;77:145–152. [PubMed] [Google Scholar]
  • 68.Odunsi K, Qian F, Matsuzaki J, Mhawech-Fauceglia P, Andrews C, Hoffman EW, Pan L, Ritter G, Villella J, Thomas B, Rodabaugh K, Lele S, Shrikant P, Old LJ, Gnjatic S. Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc Natl Acad Sci U S A. 2007;104:12837–12842. doi: 10.1073/pnas.0703342104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Odunsi K, Matsuzaki J, James SR, Mhawech-Fauceglia P, Tsuji T, Miller A, Zhang W, Akers SN, Griffiths EA, Miliotto A, Beck A, Batt CA, Ritter G, Lele S, Gnjatic S, Karpf AR. Epigenetic potentiation of NY-ESO-1 vaccine therapy in human ovarian cancer. Cancer Immunol Res. 2014;2:37–49. doi: 10.1158/2326-6066.CIR-13-0126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Samlowski WE, Leachman SA, Wade M, Cassidy P, Porter-Gill P, Busby L, Wheeler R, Boucher K, Fitzpatrick F, Jones DA, Karpf AR. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J. Clin. Oncol. 2005;23:3897–3905. doi: 10.1200/JCO.2005.06.118. [DOI] [PubMed] [Google Scholar]
  • 71.Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, Hancox A, Hong JA, Chen GA, Pishchik V, Figg WD, Murgo AJ, Steinberg SM. Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clin Cancer Res. 2006;12:5777–5785. doi: 10.1158/1078-0432.CCR-06-0669. [DOI] [PubMed] [Google Scholar]
  • 72.Srivastava P, Paluch BE, Matsuzaki J, James SR, Collamat-Lai G, Taverna P, Karpf AR, Griffiths EA. Immunomodulatory action of the DNA methyltransferase inhibitor SGI-110 in epithelial ovarian cancer cells and xenografts. Epigenetics. 2015;10:237–246. doi: 10.1080/15592294.2015.1017198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chen YT, Scanlan MJ, Venditti CA, Chua R, Theiler G, Stevenson BJ, Iseli C, Gure AO, Vasicek T, Strausberg RL, Jongeneel CV, Old LJ, Simpson AJ. Identification of cancer/testis-antigen genes by massively parallel signature sequencing. Proc Natl Acad Sci U S A. 2005;102:7940–7945. doi: 10.1073/pnas.0502583102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yang P, Huo Z, Liao H, Zhou Q. Cancer/testis antigens trigger epithelial-mesenchymal transition and genesis of cancer stem-like cells. Curr Pharm Des. 2015;21:1292–1300. doi: 10.2174/1381612821666141211154707. [DOI] [PubMed] [Google Scholar]
  • 75.Chen YT, Hsu M, Lee P, Shin SJ, Mhawech-Fauceglia P, Odunsi K, Altorki NK, Song CJ, Jin BQ, Simpson AJ, Old LJ. Cancer/testis antigen CT45: analysis of mRNA and protein expression in human cancer. Int J Cancer. 2009;124:2893–2898. doi: 10.1002/ijc.24296. [DOI] [PubMed] [Google Scholar]
  • 76.Koop A, Sellami N, Adam-Klages S, Lettau M, Kabelitz D, Janssen O, Heidebrecht HJ. Down-regulation of the cancer/testis antigen 45 (CT45) is associated with altered tumor cell morphology, adhesion and migration. Cell Commun Signal. 2013;11:41. doi: 10.1186/1478-811X-11-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Coscia F, Lengyel E, Duraiswamy J, Ashcroft B, Bassani-Sternberg M, Wierer M, Johnson A, Wroblewski K, Montag A, Yamada SD, Lopez-Mendez B, Nilsson J, Mund A, Mann M, Curtis M. Multi-level proteomics identifies CT45 as a chemosensitivity mediator and immunotherapy target in ovarian cancer. Cell. 2018;175:159–170. e116. doi: 10.1016/j.cell.2018.08.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Grizzi F, Gaetani P, Franceschini B, Di Ieva A, Colombo P, Ceva-Grimaldi G, Bollati A, Frezza EE, Cobos E, Rodriguez y Baena R, Dioguardi N, Chiriva-Internati M. Sperm protein 17 is expressed in human nervous system tumours. BMC Cancer. 2006;6:23. doi: 10.1186/1471-2407-6-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wen Y, Richardson RT, Widgren EE, O’Rand MG. Characterization of Sp17: a ubiquitous three domain protein that binds heparin. Biochem J. 2001;357:25–31. doi: 10.1042/0264-6021:3570025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Richardson RT, Yamasaki N, O’Rand MG. Sequence of a rabbit sperm zona pellucida binding protein and localization during the acrosome reaction. Dev Biol. 1994;165:688–701. doi: 10.1006/dbio.1994.1285. [DOI] [PubMed] [Google Scholar]
  • 81.O’Rand MG, Widgren EE, Fisher SJ. Characterization of the rabbit sperm membrane autoantigen, RSA, as a lectin-like zona binding protein. Dev Biol. 1988;129:231–240. doi: 10.1016/0012-1606(88)90177-7. [DOI] [PubMed] [Google Scholar]
  • 82.Yamasaki N, Richardson RT, O’Rand MG. Expression of the rabbit sperm protein Sp17 in COS cells and interaction of recombinant Sp17 with the rabbit zona pellucida. Mol Reprod Dev. 1995;40:48–55. doi: 10.1002/mrd.1080400107. [DOI] [PubMed] [Google Scholar]
  • 83.Brunette LL, Mhawech-Fauceglia PY, Ji L, Skeate JG, Brand HE, Lawrenson K, Walia S, Chiriva-Internati M, Groshen S, Roman LD, Kast WM, Da Silva DM. Validity and prognostic significance of sperm protein 17 as a tumor biomarker for epithelial ovarian cancer: a retrospective study. BMC Cancer. 2018;18:970. doi: 10.1186/s12885-018-4880-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Schutt CA, Mirandola L, Figueroa JA, Nguyen DD, Cordero J, Bumm K, Judson BL, Chiriva-Internati M. The cancer-testis antigen, sperm protein 17, a new biomarker and immunological target in head and neck squamous cell carcinoma. Oncotarget. 2017;8:100280–100287. doi: 10.18632/oncotarget.22213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Liu Y, Tian X, Jiang S, Ren X, Liu F, Yang J, Chen Y, Jiang Y. Umbilical cord blood-derived dendritic cells infected by adenovirus for SP17 expression induce antigen-specific cytotoxic T cells against NSCLC cells. Cell Immunol. 2015;298:18–24. doi: 10.1016/j.cellimm.2015.08.004. [DOI] [PubMed] [Google Scholar]
  • 86.Chiriva-Internati M, Wang Z, Salati E, Bumm K, Barlogie B, Lim SH. Sperm protein 17 (Sp17) is a suitable target for immunotherapy of multiple myeloma. Blood. 2002;100:961–965. doi: 10.1182/blood-2002-02-0408. [DOI] [PubMed] [Google Scholar]
  • 87.Lim SH, Wang Z, Chiriva-Internati M, Xue Y. Sperm protein 17 is a novel cancer-testis antigen in multiple myeloma. Blood. 2001;97:1508–1510. doi: 10.1182/blood.v97.5.1508. [DOI] [PubMed] [Google Scholar]
  • 88.Li Z, Li W, Meklat F, Wang Z, Zhang J, Zhang Y, Lim SH. A yeast two-hybrid system using Sp17 identified Ropporin as a novel cancer-testis antigen in hematologic malignancies. Int J Cancer. 2007;121:1507–1511. doi: 10.1002/ijc.22842. [DOI] [PubMed] [Google Scholar]
  • 89.Straughn JM Jr, Shaw DR, Guerrero A, Bhoola SM, Racelis A, Wang Z, Chiriva-Internati M, Grizzle WE, Alvarez RD, Lim SH, Strong TV. Expression of sperm protein 17 (Sp17) in ovarian cancer. Int J Cancer. 2004;108:805–811. doi: 10.1002/ijc.11617. [DOI] [PubMed] [Google Scholar]
  • 90.Li FQ, Han YL, Liu Q, Wu B, Huang WB, Zeng SY. Overexpression of human sperm protein 17 increases migration and decreases the chemosensitivity of human epithelial ovarian cancer cells. BMC Cancer. 2009;9:323. doi: 10.1186/1471-2407-9-323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nakazato T, Kanuma T, Tamura T, Faried LS, Aoki H, Minegishi T. Sperm protein 17 influences the tissue-specific malignancy of clear cell adenocarcinoma in human epithelial ovarian cancer. Int J Gynecol Cancer. 2007;17:426–432. doi: 10.1111/j.1525-1438.2007.00815.x. [DOI] [PubMed] [Google Scholar]
  • 92.Gao Q, Xiang SD, Wilson K, Madondo M, Stephens AN, Plebanski M. Sperm protein 17 expression by murine epithelial ovarian cancer cells and its impact on tumor progression. Cancers (Basel) 2018;10:276. doi: 10.3390/cancers10080276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chiriva-Internati M, Yu Y, Mirandola L, Jenkins MR, Chapman C, Cannon M, Cobos E, Kast WM. Cancer testis antigen vaccination affords long-term protection in a murine model of ovarian cancer. PLoS One. 2010;5:e10471. doi: 10.1371/journal.pone.0010471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Song JX, Li FQ, Cao WL, Jia X, Shi LN, Lu JF, Ma CF, Kong QQ. Anti-Sp17 monoclonal antibody-doxorubicin conjugates as molecularly targeted chemotherapy for ovarian carcinoma. Target Oncol. 2014;9:263–272. doi: 10.1007/s11523-013-0293-6. [DOI] [PubMed] [Google Scholar]
  • 95.Vermeij R, Daemen T, de Bock GH, de Graeff P, Leffers N, Lambeck A, ten Hoor KA, Hollema H, van der Zee AG, Nijman HW. Potential target antigens for a universal vaccine in epithelial ovarian cancer. Clin Dev Immunol. 2010;2010:891505. doi: 10.1155/2010/891505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kanduc D. Oligopeptides for immunotherapy approaches in ovarian cancer treatment. Curr Drug Discov Technol. 2019;16:285–289. doi: 10.2174/1570163815666180525071740. [DOI] [PubMed] [Google Scholar]
  • 97.Xiang SD, Gao Q, Wilson KL, Heyerick A, Plebanski M. Mapping T and B cell epitopes in sperm protein 17 to support the development of an ovarian cancer vaccine. Vaccine. 2015;33:5950–5959. doi: 10.1016/j.vaccine.2015.07.094. [DOI] [PubMed] [Google Scholar]
  • 98.Mattila JP, Mirandola L, Chiriva-Internati M. Development of a M cell-targeted microparticulate platform, BSK02, for oral immunization against the ovarian cancer antigen, sperm protein 17. J Biomed Mater Res B Appl Biomater. 2019;107:29–36. doi: 10.1002/jbm.b.34092. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES