Skip to main content
Immunotherapy logoLink to Immunotherapy
. 2018 Jun 20;10(9):769–778. doi: 10.2217/imt-2017-0179

Cancer testis antigens as immunogenic and oncogenic targets in breast cancer

Abeer M Mahmoud 1,1,2,2,*
PMCID: PMC6462849  PMID: 29926750

Abstract

Breast cancer cells frequently express tumor-associated antigens that can elicit immune responses to eradicate cancer. Cancer-testis antigens (CTAs) are a group of tumor-associated antigens that might serve as ideal targets for cancer immunotherapy because of their cancer-restricted expression and robust immunogenicity. Previous clinical studies reported that CTAs are associated with negative hormonal status, aggressive tumor behavior and poor survival. Furthermore, experimental studies have shown the ability of CTAs to induce both cellular and humoral immune responses. They also demonstrated the implication of CTAs in promoting cancer cell growth, inhibiting apoptosis and inducing cancer cell invasion and migration. In the current review, we attempt to address the immunogenic and oncogenic potential of CTAs and their current utilization in therapeutic interventions for breast cancer.

Keywords: : breast cancer, cancer immunology, cancer testis antigens, immunotherapy, tumor-associated antigens


Breast cancer is the most common cancer affecting women in the USA. In breast cancer, genetic and epigenetic events can generate unique antigens that are expressed in tumors as they progress. These antigens are known as tumor-associated antigens (TAAs), and they either induce an expansion of CD8+ cytotoxic T lymphocytes (CTLs) that are capable of rejecting tumor cells or activate B lymphocytes and subsequently elicit a humoral response in the form of tumor-specific antibodies [1]. Breast cancers frequently express TAAs on their surface that may potentially serve as future targets for immunotherapy. TAA-based immunotherapy could be an alternative treatment strategy for patients with hormone receptor-negative breast cancer who lack estrogen receptors, and Her2. The latter are usual targets for therapies such as tamoxifen and trastuzumab, and their absence leaves chemotherapy as the only treatment option for this category of breast cancer patients. These TAAs span a spectrum of immunogenic capacity, TAAs with stronger immunogenic potential is more likely to elicit an adequate immune response. Cancer-testis antigens (CTAs) are a category of TAAs that have received particular attention because of their high immunogenicity [2]. CTA expression is usually restricted to the testis and is otherwise silenced by promoter hypermethylation [3]. In several cancers, however, CTA genes are frequently demethylated and consequently re-expressed, with potentially significant alteration of the host immune response [4]. This cancer-restricted pattern of expression, together with their strong immunogenicity, identifies CTAs as ideal targets for cancer immunotherapy and could prompt the development of CTA-based vaccine treatment that should help patients with limited treatment options such as those with triple-negative breast cancer.

CTA epitopes are presented on the surface of cancer cells in association with human leukocyte antigen (HLA) class I molecules. The antigen-specific T-cell receptor on the surface of CD8+ CTLs recognizes the epitope/HLA complex resulting in the killing of the antigen-bearing tumor cells. With active specific immunotherapy, CTAs coupled to appropriate adjuvant can elicit robust antitumor responses. The potential advantages of therapeutic cancer vaccines are that they can augment an established immunogenic response to the tumor, target specific tumor antigens and elicit immunologic memory to prevent recurrence of the tumor.

In addition to immunogenicity, CTAs have oncogenic functions including support of tumor growth (i.e., SSX and CAGE), evasion of apoptosis (i.e., MAGE A/B/C, GAGE and PAGE4), and induction of invasion and metastasis (i.e., MAGE C2, CAGE, GAGE, XAGE and CT45A1). This novel insight into CTA function will help develop therapeutic strategies that combine immune targeting of oncogenic CTAs with conventional cytotoxic therapies. Accordingly, a significant effort is required to identify optimal therapeutic targets that are highly immunogenic, HLA-presented, cancer-specific and frequently expressed in breast cancer tissues, which make them promising candidates for cancer immunotherapy.

Expression of CTAs in breast cancer

MAGE-A and NY-ESO-1 are the prototypes of CTAs and among the first discovered TAAs that elicit natural cellular immunity in cancers such as melanoma and ovarian, bladder and lung cancer [5,6]. Yet, their expression in breast cancer has been discovered relatively recently. There are more than 100 identified CTAs to date and among those emerging as most relevant to breast cancer are MAGE family (MAGE-A, MAGE-B, MAGE-C), GAGE family (GAGE-A, GAGE-B, PAGE, XAGE), NY-ESO-1, BAGE and BORIS.

Previous studies have confirmed the expression of several CTAs in breast cancer tissues. On top of the well-characterized CTAs are MAGE and NY-ESO-1. The mRNA and protein expression of MAGE-A9, -A10 and -A11 was detected in invasive ductal carcinoma samples [7,8], and their expression was significantly higher than the adjacent noncancerous tissues. A study by Bandić et al. [4] showed that MAGE-A4 and NY-ESO-1 proteins were expressed in 74 and 40%, respectively, in a cohort of 81 Croatian women with invasive breast cancer. Measuring CTA protein expression in a prospective cohort of 100 German women with primary breast cancer has detected an expression of GAGE in 26%, NY-ESO-1 in 13%, MAGE-3 in 11% and MAGE-4 in only 4% of patients [9]. In this study MAGE and NY-ESO-1 were not the predominantly expressed CTAs. Instead, SCP-1 and SSX-4 were expressed most frequently (both 65%), followed by HOM-TES-85/CT-8 (47%). In a cohort of 82 breast cancer patients, Taylor et al. [10] have found that more than two-thirds of tumors expressed, at the mRNA level, at least one of the following CTAs: CTAG1, BAGE1 and MAGEA-10, all of which are known to be potent immunogenic targets of cytotoxic-T-lymphocyte. SPAG, a recently characterized CTA, was reported to be expressed in over 60% of breast cancer cases [11]. Similarly, the expression of several other CTAs were detected in breast cancer tissues such as: LUZP4 [12], ATPase Family, ATAD2 [13,14], RHOXF2, ODF4 [15], SPANX-A/C/D [16], FBXO39, TDRD4 [17] and WBP2NL [18].

An additional aspect to consider, when we discuss CTA expression in breast cancer, is the reliability of the methods used to characterize and measure CTAs in cancer tissues. Quantitative polymerase chain reaction (qRT-PCR) is a simple and sensitive method to characterize CTA profiles of individual tumors especially if tissue material is limited. However, using mRNA as a surrogate parameter of CTA expression assumes that the level of mRNA correlates with the level of the protein it encodes. Unfortunately, this is not true for many CTAs which arise from several steps of processing such as proteasomal degradation and post-translational modifications that take place prior to antigen presentation. Loss of components of the antigen-processing pathway, which has been reported as an immune evasion mechanism in some cancers [19], might contribute to the lack of correlation between mRNA and protein levels of some CTAs such as MAGE-1 [20]. Thus, direct detection and quantification of CTA proteins is expected to be one step closer to the final product that is expressed on cancer cell surface and exert antigenic properties; yet, this approach is not without limitations. One of the biggest challenges that face CTA protein characterization is the less than optimal specificity and sensitivity of most of the commercially available antibodies. Furthermore, post-translational modifications of CTAs, such as protein glycosylation, may modify the ability of the antibodies to recognize and/or bind to their corresponding antigens [20]. In the recent years, mass spectrometry-based methods of proteome analysis have gained wide acceptance in research settings since they are more comprehensive, sensitive and reproducible and hold considerable promise for the discovery of novel TAAs [21,22]. However, it is important to be aware of the limitations of TAA profiling by mass spectrometry which include the need for large amounts of tissue to detect antigens that exist at low concentration. Also, other factors such as sample collection, storage, processing and purification may produce proteomic artifacts that could mask those representing of cancer. Collectively, the large number of CTAs reported by several studies using heterogenous sets of cancer tissues and cell models, makes it difficult to verify to what extent the inconsistency in the employed methodology has contributed to the existing discrepancy in literature. Therefore, there is a heightened need for integrated genetic and proteomic approaches that are aided by bioinformatic analysis to better characterize the antigenic profiles in breast cancer.

The biological role of CTAs in breast cancer development and progression remains poorly understood. However, a growing experimental evidence indicates that their expression could have a role in tumorigenesis via regulating cancer cell proliferation and apoptosis as well as their invasive and metastatic priorities. Thus, studying the immunogenicity of these CTAs may facilitate targeting them via immunotherapy, which will benefit a broad group of breast cancer patients especially those with negative hormonal tumors who are not eligible for hormonal treatment. Furthermore, identifying the oncogenic properties for CTAs may help develop a robust, synergistic approach that combines immunotherapy with conventional cytotoxic therapies. In the following sections, we will discuss the immunogenic and oncogenic roles of CTAs in breast cancer.

CTAs as immunogenic targets in breast cancer

CTAs are excellent targets for cancer immunotherapy such as cancer vaccination, adoptive T-cell transfer with a chimeric T-cell receptor or as an adjuvant treatment with the conventional cancer cytotoxic drugs. CTA-based immunotherapy represents a highly synergistic approach that has a significant potential of improving patient's survival because they are more specific, lower in toxicity and have potential to eradicate residual disease and metastases and the immunological memory induced by cancer vaccines can prevent tumor recurrence [23,24]. CTA-based vaccination has yielded favorable immunological and clinical responses in some neoplastic lesions such as melanoma and ovarian, bladder and lung cancer (reviewed in Gjerstorff et al. [25] and Fratta et al. [26]). It was recently reported that in MAGRIT Phase III Trial, MAGE-A3-based vaccine did not increase disease-free survival in patients with non-small-cell lung cancer (NSCLC) compared with the placebo-receiving patients [27]. However, it is worth mentioning that the aggressive nature of the NSCLC and accordingly, the short survival of patients with NSCLC might constitute a great obstacle in assessing the long-term efficacy of CTA-based vaccines. Furthermore, the strongly immunosuppressive tumoral environment in NSCLC and the use of a vaccine that is based on one CTA isoform (MAGE-3A) might have contributed to the lack of clinical benefits in this trial.

MAGE-A and NY-ESO-1 are on the top of the list of CTAs that have been tested in a variety of clinical settings [28] and are currently being investigated in many cancer vaccination trials, some of which have provided encouraging outcomes [29,30]. For example, patients with melanoma and ovarian cancer showed significant induction of both humoral and cellular immune responses and increased survival in response to NY-ESO-1 and MAGE-A vaccines and adoptive T-cell transfer [25,31]. Dendritic cell-based, tumor-associated antigen peptide-based, and whole cell-based vaccines are currently tested approaches of cancer immunotherapy [32].

Despite these promising results of CTA-based immunotherapy in many cancers, the reported vaccination trials in breast cancer patients are very few which might indicate either an actual lack of conducting CTA-based vaccination trials in breast cancer or null results that were not published. After thorough research, we came across two trials. The first by Janosky et al. reported two cases of metastatic breast cancer with immune response and local remission of skin metastasis after in situ vaccination with MAGE-specific T-cells [33]. Another clinical trial (Phase I) that was published as an abstract in the ASCO meeting, 2012 by Fujita et al. [34] demonstrated that combining five kinds of CTA peptide vaccination (CDCA1, URLC10, KIF20A, DEPDC1 and MPHOSPH1) in nine patients with metastatic and advanced breast cancer induced local immunological response at site of injection in 44% and systemic increase in cytotoxic T-cells in 78 and 60% of patients who completed at least two cycles of the treatment demonstrated significant tumor regression. Currently, there are few ongoing clinical trials that investigate the effect of MAGE-A or NY-ESO-1-based vaccines on patients with primary or metastatic cancers that express these antigens including breast cancer [35,36].

The therapeutic potential of breast cancer CTAs has also been shown in a variety of in vitro and mechanistic studies. A study by Liu et al. [37] have identified two PLAC1 peptides (p28 and p31) and their synthetic analogs (p28–1Y9V, p31–1Y2L) as high-affinity peptides, with strong binding affinity and stability to the HLA-A*0201 molecule. These peptides induced a potent cytotoxic T-cell response that could lyse MCF-7 cells. Another peptide (p101–111) that was derived from SSX2 antigen showed dual immunogenicity for both helper and cytotoxic T-cells resulting in both humoral and cellular-mediated immune responses in vitro [38].

There is growing evidence that breast cancer is immunogenic. However, it appears that the spontaneous immune responses are unable to control tumor growth and, thus, further research is needed to enhance the host immune responses against breast cancer by therapeutic interventions. To improve the chance of women with breast cancer to benefit from the emerging immunotherapeutic strategies, it is necessary to understand the pattern of expression of CTAs in breast cancer, identify the most prevalent and immunogenic subtypes and exploring their function and relation to breast cancer outcomes.

The family of CTAs is expanding rapidly, and new members are continuously added to the list. The expression of these CTAs varies widely among different studies; however, a common and consistent finding is that no individual CTA is expressed in all breast cancers and that CTAs are always expressed in heterogeneous combinations in different individuals. This heterogeneity is expected to minimize the efficacy of standardized immunotherapy regimens which entails introducing the next generation of immunotherapy approaches that take into consideration patient's CTA repertoire. Also, this indicates the importance of developing a polyvalent vaccine that covers a large number of the most common and immunogenic CTAs in breast cancer tissues; a strategy that is already in clinical development for the treatment of melanoma [39].

T-cell responses, particularly the one mediated via the CD8+ CTL, are believed to be the most important element of immune responses that target CTAs in breast cancers. Previous studies have identified CTL responses against the most prevalent three CTAs: CTAG1, BAGE1 and MAGEA10 in breast cancers [40–42]. A vaccine that targets these three antigens would cover more than two-thirds of breast tumors. HLA-presented peptide epitopes of these CTAs provide potential targets for a clinically relevant cytotoxic T-cell response. Therefore, therapeutic vaccines based on the naturally presented HLA-CTA peptide repertoire might induce tangible effects on tumor eradication and patient survival. CTAG1 is the most studied of the three CTAs, and its peptides are presented by several HLA molecules including HLA A2, HLA A31, HLA B7, HLA B35, HLA B51, HLA Cw3 and HLA Cw6, which are common HLA alleles in the general population [43]. MAGEA10 is also a well-studied CTA, and its peptides are presented by HLA A2 and HLA B53 [44]. BAGE1, which is expressed in more than 10% of breast cancers, is presented by HLA Cw16, however, bioinformatic models identified potential high-affinity of BAGE peptide epitopes for several other HLA molecules, and even a larger number of potential high-affinity epitopes were reported for MAGEA10 [41,45].

CTAs as oncogenic targets in breast cancer

Several studies reported the activation of CTAs in breast cancer cells demonstrating their role in cell cycle regulation, proliferation, apoptosis and promoting invasion in breast cancer. MAGE, a well-characterized CTA, has been shown to play a significant role in breast cancer cell survival via inhibiting P53, an important tumor suppressor gene, thereby increasing cell proliferation and reducing apoptosis [46]. It has been suggested that MAGE-A inhibits P53 by binding to the P53 core domain and regulating its transcriptional activity or by modulating p53 acetylation through the recruitment of HDAC3 [47]. In addition to regulating p53 directly, MAGE-A can modify the function of MDM2 and MDM4 which are important negative regulators of the p53 gene [48]. GPAT2 is another example of CTAs which mechanism of action has been studied in breast cancer cells (i.e., MDA-MB-231 and MCF7). Silencing of GPAT2 reduced MDA-MB-231 cell proliferation, anchorage-independent growth, migration and tumorigenicity [49]. Similarly, in vitro and in vivo xenograft studies showed that silencing of the SPAG9 reduced growth and invasive potential of breast cancer cells via upregulating p21 along with other pro-apoptotic molecules and epithelial markers and downregulating signaling molecules that are involved in cell cycle progression and epithelial–mesenchymal transition (EMT) [50]. TSP50, a newly discovered TSA, has also been shown to be involved in breast cancer cell proliferation and cell cycle progression via inhibiting P53 signaling, inducing cyclin D and cyclin E, and promoting G1-S transition [51].

Other CTAs have been shown to promote breast cancer cell invasion and metastasis and their expression in breast cancer tissues correlated with a higher risk of organ metastasis. The ability of these CTAs to initiate EMT in breast cancer cells provides a plausible explanation for their association with enhanced invasion and metastasis. On the top of this category of CTAs are CT45A1, TSP50, MAGEC2, SPANX-A/C/D and CTAG2. Shang et al. [52] have shown that CT45A1 overexpression in MCF7 breast cancer cells is associated with upregulation of several essential EMT genes such as TWIST1, ALDH1A1 and the tyrosine-protein kinase KIT. MAGE-C2 is also capable of inducing EMT phenotype in MCF7 breast cancer cells by binding to a transcriptional repressor, KAP1, which plays a critical role in promoting cadherin switching and acquiring mesenchymal cell molecular patterns [53,54]. Similarly, TSP50 has been shown to induce EMT in breast cancer cells, yet it is also capable of activating a group of matrix metalloproteinases that degrade the extracellular matrix and facilitate tumor cell migration and invasion [55]. SPANX-A/C/D and CTAG2 are also involved in breast cancer cell migration and metastatic potential, and the underlying mechanism is thought to be a consequence of CTA-induced modifications in Lamin A/C which subsequently results in remodeling of extracellular matrix, reduction in E-cadherin and loss of cell adherence [16].

The expression of CTAs varied widely among breast cancer patients and some studies reported a trend toward having higher risk features such as advanced histological grade and clinical stage, lymph node metastasis and hormonal receptor negative status among cases with high CTA expression [10,56–58]. Also, CTA expression correlated with distinct molecular patterns which might indicate their relevance as prognostic biomarkers in breast cancer. For example, NY-ESO-1 expression that was measured in a cohort of 1234 breast cancer patients correlated with reduced disease-free survival. NY-ESO-1 expression was also associated with a higher level of tumor-infiltrating lymphocytes which suggests that it is a strong immunogenic CTA that could serve as an ideal immunotherapeutic target [59]. Similarly, MAGE expression in breast cancer tissues correlated with advanced tumor stage, lymph node metastasis, larger tumor size, tumor recurrence and poor overall survival. Also, MAGE was significantly associated with ER- and PR-negative status. These studies indicated that MAGE is a potential novel prognostic marker in breast cancer. A study conducted at Ludwig Institute for Cancer Research, NY reported higher levels of MAGE-A and NY-ESO-1 in ER-negative (47%) than ER-positive (8%) breast cancer cases and in brain metastasis (66%) than primary tumors [57]. Lee et al. [59] have shown that NY-ESO-1 was mainly expressed in triple negative breast cancer samples (TNBC). Curigliano et al. have confirmed immunohistochemically that both MAGE-A and NY-ESO-1 are highly expressed in TNBC compared with ER-positive tumors in a small cohort of 100 invasive breast cancer samples [60]. In another study by Chen et al. [58], analyzed the protein expression of several CTAs including MAGE-A, NY-ESO-1, CT45, GAGE and SAGE1 in 454 invasive ductal carcinomas. They reported more frequent expression of CTAs in ER-negative than ER positive cancers; however, HER2 status had no consistent effect on CTAs expression. In this study, MAGE-A and NY-ESO-1 were also highly expressed in TNBC, showing similar results to what has been reported by Curigliano et al. [60]. Similarly, an earlier study by Grigoriadis et al. [61] demonstrated a higher expression frequency of MAGEA and NY-ESO-1 as well as the CTAs that are located on the X chromosome (CTA-X) in ER- and PR-negative breast cancer cell lines and primary breast carcinomas. Collectively, these findings indicate that CT antigens expression is closely related to the ER status. Other CTAs such as HORMAD1, CXorf61, ACTL8 and PRAME were found to be highly expressed in the breast cancers that have basal cell properties [62]. Furthermore, there are many other CTAs that predict invasion and metastasis of breast cancer such as ATAD2 [13], GPAT2 [49], NY-BR-1 [63] and BORIS [64] (Table 1 and Figure 1).

Table 1. . Expression and function of cancer-testis antigens in breast cancer.

CTAs Breast cancer clinical specimens/cell lines Breast cancer molecular subtypes CTA oncogenic Function Ref.
ACTL8 Breast cancer tissues TNBC-Basal-like Survival and evasion of apoptosis [62]

ALDH1A1 Breast cancer tissues, MDA-MB-468 and SUM159 NS Metastasis, invasion and stem cell proliferation [66–68]

ATAD2 Breast cancer tissues ER positive Growth, invasion and metastasis [13,69]

BAGE Breast cancer tissues TNBC Invasion and metastasis [10]

BORIS (CTCFL) Breast cancer tissues MDA-MB-231, and MCF7 ER and PR positive Survival, transcriptional regulation and epigenetic modifications [70,71]

CAGE Breast cancer tissues NS Growth, survival and angiogenesis [72]

CDCA1 MCF7, MDA-MB-231, MDA-MB-453, ZR-75–1 and HTB-19 NS Proliferation and mitosis [73]

CT45A1 Breast cancer tissues and MCF-7 NS Modulation of cell morphology, adherence and motility, metastasis and invasion [52,74]

CTAG1 Breast cancer tissues and SUM159 TNBC Invasion and metastasis [10,16]

FBXO39 Breast cancer tissues, MCF-7 and MDA-MB-231 NS Tumorigenesis [17]

GAGE Breast cancer tissues TNBC-Basal-like Survival, invasion, metastasis, epigenetic modifications and stem cell proliferation [75]

GPAT2 MDA-MB-231 NS Proliferation, survival, anchorage independent growth and migration [49]

HORMAD1 MDA-MB-468 TNBC-Basal-like Genomic instability and hormonal resistance [62,76]

KIF20A Breast cancer tissues and MCF-7 ER positive Growth, survival and hormonal resistance [77]

LUZP4 Breast cancer tissues Luminal A, Luminal B and Basal-like Codifies a leucine zipper protein that is involved in mRNA exporting in cancer cells [12]

MAGE-A Breast cancer tissues, MCF-7 and MDA-MB-231 TNBC Survival, invasion, metastasis and epigenetic modifications [7,8,75]

NY-ESO-1 Breast cancer tissues TNBC Growth and metastasis [56,59]

ODF4 Breast cancer tissues and MDA-MB-231 Her2-enriched Proliferation and transcriptional activity [15]

PAGE4 Breast cancer tissues NS Survival and evasion of apoptosis [78]

PIWIL2 Breast cancer tissues and MDA-MB-231 NS Survival and evasion of apoptosis [79]

PLAC1 Breast cancer tissues, MCF-7 and BT-549 NS Motility, migration, invasion and G1-S cell cycle progression [80]

PRAME Breast cancer tissues TNBC-Basal-like Proliferation, differentiation and Dominant repressor of retinoic acid Signaling [62,81]

RHOXF2 Breast cancer tissues, MCF-7 and MDA-MB-231 Her2-enriched Proliferation and transcriptional activity [15]

SPAG Breast cancer tissues and MDA-MB-231 TNBC Growth, metastasis and invasion [11,82]

SPANX-A/C/D Breast cancer tissues and SUM159 TNBC Invasion and metastasis [16,75]

SSX1/2/4 Breast cancer tissues and MCF-7 NS Proliferation and genomic instability [83]

TDRD4 Breast cancer tissues and MDA-MB-231 NS Tumorigenesis [17]

TSP50 Breast cancer tissues and MDA-MB-231 NS Threonine protease that performs a critical role in tumorigenesis [84]

WBP2NL Breast cancer tissues, MCF-7 and MDA-MB-231 NS Proliferation and tumorigenesis [18]

XAGE Breast cancer tissues and MCF-7 NS Invasion and metastasis [75,78]

CTA: Cancer-testis antigen; NS: Not specified; TNBC: Triple negative breast cancer.

Figure 1. . Schematic illustration of the immunogenic versus oncogenic functions of cancer-testis antigens and their potential clinical applications.

Figure 1. 

CTAs that are expressed on the surface of breast cancer cells may have immunogenic and oncogenic properties. The strongly immunogenic CTAs may induce an expansion of CD8+ T cells that are capable of rejecting tumor cells (cellular immunity) or activate B cells that elicit a humoral response in the form of tumor-specific antibodies. At the same time, CTAs may demonstrate oncogenic capabilities in breast cancer. They have been shown to be involved in steps of tumorigenesis that include induction of tumor growth and angiogenesis, evasion of apoptosis, enhancement of tumor migration (invasion and metastasis) and generation of genomic instability. Accordingly, CTAs are excellent candidates for synergistic approaches that combine both immunotherapies with conventional cytotoxic therapies.

CTA: Cancer-testis antigen.

Future perspective

This different subtypes and frequencies of CTA expression in breast cancer could be a major source of discouragement of vaccination trials in this type of cancer. In general, heterogeneity in CTA expression and immunogenicity and the inherent immune evasion ability of some cancers are the most common challenges that face the development of an effective immunotherapy. However, these obstacles should not stand in the way of developing effective breast cancer vaccines or immunotherapies that might be the only hope for some women with hormone-resistant breast cancers. New strategies to circumvent these challenges in the broad field of immunotherapy are being currently investigated. Recently, it has been shown that designing vaccines that contain peptides from several CTAs is a successful strategy to overcome the problem of CTA heterogeneity among individuals and prevent immune evasion that might occur through selective loss of single target antigens [65]. Also, there is a need for the formulation of single patient-tailored vaccines which would comprise a significant part of the individual patient's CTA repertoire, including strongly immunogenic unique tumor antigens. More research is needed for a better understanding of the involvement of CTAs in the mechanisms of antitumor immune responses that mediate the clinical outcome of breast cancer. It will help to comprehend the molecular basis of immune recognition and tumor immunogenicity as well as to reveal the molecular characterization and functional heterogeneity of tumor-associated immune cells. Also, a significant effort is required to elucidate disparities in the biology, response to immunotherapy and outcomes seen in women with breast cancer from different racial/ethnic groups. Thus, future studies should focus on regimens integrating observational studies of CTA expression and immune responses in clinical samples, correlational studies with outcomes such as recurrence and survival and finally mechanistic studies that investigate CTA function and immunogenicity.

Executive summary.

  • The cancer-restricted pattern of expression of cancer-testis antigens (CTAs) and their strong immunogenicity identifies them as promising candidates for cancer immunotherapy.

  • Previous studies reported the expression of CTAs in breast cancer tissues mainly MAGE and NY-ESO-1.

  • Theoretically, more than two-thirds of breast cancer patient would benefit from CTA-based vaccines, however, results from CTA-based vaccination trials were not as aspired.

  • In addition to their immunogenic nature, CTAs have demonstrated a role in cancer cell growth and invasive properties.

Footnotes

Financial & competing interests disclosure

This work was supported by the following funding source: NIH-1K99HL140049-01 (AMM). The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • 1.Lim SH, Zhang Y, Zhang J. Cancer-testis antigens: the current status on antigen regulation and potential clinical use. Am. J. Blood Res. 2012;2(1):29–35. [PMC free article] [PubMed] [Google Scholar]
  • 2.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]
  • 3.Glazer CA, Smith IM, Ochs MF, et al. Integrative discovery of epigenetically derepressed cancer testis antigens in NSCLC. PLoS One. 2009;4(12):e8189. doi: 10.1371/journal.pone.0008189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bandic D, Juretic A, Sarcevic B, et al. Expression and possible prognostic role of MAGE-A4, NY-ESO-1, and HER-2 antigens in women with relapsing invasive ductal breast cancer: retrospective immunohistochemical study. Croat. Med. J. 2006;47(1):32–41. [PMC free article] [PubMed] [Google Scholar]
  • 5.Davis ID, Chen W, Jackson H, et al. Recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4(+) and CD8(+) T cell responses in humans. Proc. Natl Acad. Sci. USA. 2004;101(29):10697–10702. doi: 10.1073/pnas.0403572101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Jager E, Nagata Y, Gnjatic S, et al. Monitoring CD8 T cell responses to NY-ESO-1: correlation of humoral and cellular immune responses. Proc. Natl Acad. Sci. USA. 2000;97(9):4760–4765. doi: 10.1073/pnas.97.9.4760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Xu X, Tang X, Lu M, et al. Overexpression of MAGE-A9 predicts unfavorable outcome in breast cancer. Exp. Mol. Pathol. 2014;97(3):579–584. doi: 10.1016/j.yexmp.2014.11.001. [DOI] [PubMed] [Google Scholar]
  • 8.Lian Y, Sang M, Ding C, et al. Expressions of MAGE-A10 and MAGE-A11 in breast cancers and their prognostic significance: a retrospective clinical study. J. Cancer Res. Clin. Oncol. 2012;138(3):519–527. doi: 10.1007/s00432-011-1122-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mischo A, Kubuschok B, Ertan K, et al. Prospective study on the expression of cancer testis genes and antibody responses in 100 consecutive patients with primary breast cancer. Int. J. Cancer. 2006;118(3):696–703. doi: 10.1002/ijc.21352. [DOI] [PubMed] [Google Scholar]
  • 10.Taylor M, Bolton LM, Johnson P, Elliott T, Murray N. Breast cancer is a promising target for vaccination using cancer-testis antigens known to elicit immune responses. Breast Cancer Res. 2007;9(4):R46. doi: 10.1186/bcr1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Silina K, Zayakin P, Kalnina Z, et al. Sperm-associated antigens as targets for cancer immunotherapy: expression pattern and humoral immune response in cancer patients. J. Immunother. 2011;34(1):28–44. doi: 10.1097/CJI.0b013e3181fb64fa. [DOI] [PubMed] [Google Scholar]
  • 12.De Anda-Jauregui G, Velazquez-Caldelas TE, Espinal-Enriquez J, Hernandez-Lemus E. Transcriptional network architecture of breast cancer molecular subtypes. Front. Physiol. 2016;7:568. doi: 10.3389/fphys.2016.00568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Caron C, Lestrat C, Marsal S, et al. Functional characterization of ATAD2 as a new cancer/testis factor and a predictor of poor prognosis in breast and lung cancers. Oncogene. 2010;29(37):5171–5181. doi: 10.1038/onc.2010.259. [DOI] [PubMed] [Google Scholar]
  • 14.Kalashnikova EV, Revenko AS, Gemo AT, et al. ANCCA/ATAD2 overexpression identifies breast cancer patients with poor prognosis, acting to drive proliferation and survival of triple-negative cells through control of B-Myb and EZH2. Cancer Res. 2010;70(22):9402–9412. doi: 10.1158/0008-5472.CAN-10-1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kazemi-Oula G, Ghafouri-Fard S, Mobasheri MB, Geranpayeh L, Modarressi MH. Upregulation of RHOXF2 and ODF4 expression in breast cancer tissues. Cell J. 2015;17(3):471–477. doi: 10.22074/cellj.2015.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Maine EA, Westcott JM, Prechtl AM, Dang TT, Whitehurst AW, Pearson GW. The cancer-testis antigens SPANX-A/C/D and CTAG2 promote breast cancer invasion. Oncotarget. 2016;7(12):14708–14726. doi: 10.18632/oncotarget.7408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Seifi-Alan M, Shamsi R, Ghafouri-Fard S, et al. Expression analysis of two cancer-testis genes, FBXO39 and TDRD4, in breast cancer tissues and cell lines. Asian Pac. J. Cancer Prev. 2014;14(11):6625–6629. doi: 10.7314/apjcp.2013.14.11.6625. [DOI] [PubMed] [Google Scholar]
  • 18.Nourashrafeddin S, Dianatpour M, Aarabi M, Mobasheri MB, Kazemi-Oula G, Modarressi MH. Elevated expression of the testis-specific gene WBP2NL in breast cancer. Biomark. Cancer. 2015;7:19–24. doi: 10.4137/BIC.S19079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kaklamanis L, Leek R, Koukourakis M, Gatter KC, Harris AL. Loss of transporter in antigen processing 1 transport protein and major histocompatibility complex class I molecules in metastatic versus primary breast cancer. Cancer Res. 1995;55(22):5191–5194. [PubMed] [Google Scholar]
  • 20.Kayser S, Watermann I, Rentzsch C, Weinschenk T, Wallwiener D, Guckel B. Tumor-associated antigen profiling in breast and ovarian cancer: mRNA, protein or T cell recognition? J. Cancer Res. Clin. Oncol. 2003;129(7):397–409. doi: 10.1007/s00432-003-0445-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Weinschenk T, Gouttefangeas C, Schirle M, et al. Integrated functional genomics approach for the design of patient-individual antitumor vaccines. Cancer Res. 2002;62(20):5818–5827. [PubMed] [Google Scholar]
  • 22.Chahal FC, Entwistle J, Glover N, Macdonald GC. A targeted proteomic approach for the identification of tumor-associated membrane antigens using the ProteomeLab PF-2D in tandem with mass spectrometry. Biochem. Biophys. Res. Commun. 2006;348(3):1055–1062. doi: 10.1016/j.bbrc.2006.07.187. [DOI] [PubMed] [Google Scholar]
  • 23.Soliman H. Developing an effective breast cancer vaccine. Cancer Control. 2010;17(3):183–190. doi: 10.1177/107327481001700307. [DOI] [PubMed] [Google Scholar]
  • 24.Sabel MS, Nehs MA. Immunologic approaches to breast cancer treatment. Surg. Oncol. Clin. N. Am. 2005;14(1):1–31. doi: 10.1016/j.soc.2004.07.003. [DOI] [PubMed] [Google Scholar]
  • 25.Gjerstorff MF, Andersen MH, Ditzel HJ. Oncogenic cancer/testis antigens: prime candidates for immunotherapy. Oncotarget. 2015;6(18):15772–15787. doi: 10.18632/oncotarget.4694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fratta E, Coral S, Covre A, et al. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol. Oncol. 2011;5(2):164–182. doi: 10.1016/j.molonc.2011.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Vansteenkiste JF, Cho BC, Vanakesa T, et al. 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(6):822–835. doi: 10.1016/S1470-2045(16)00099-1. [DOI] [PubMed] [Google Scholar]
  • 28.Gjerstorff MF, Burns J, Ditzel HJ. Cancer-germline antigen vaccines and epigenetic enhancers: future strategies for cancer treatment. Exp. Opin. Biol. Ther. 2010;10(7):1061–1075. doi: 10.1517/14712598.2010.485188. [DOI] [PubMed] [Google Scholar]
  • 29.US National Library of Medicine. MAGE-A in Cancer. https://clinicaltrials.gov/ct2/results?term=MAGE&cond=Cancer
  • 30.US National Library of Medicine. NY-ESO-1 in Cancer. https://clinicaltrials.gov/ct2/results?term=NY-ESO-1&cond=Cancer
  • 31.Dhodapkar MV, Sznol M, Zhao B, et al. Induction of antigen-specific immunity with a vaccine targeting NY-ESO-1 to the dendritic cell receptor DEC-205. Sci. Transl. Med. 2014;6(232):232ra251. doi: 10.1126/scitranslmed.3008068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ghafouri-Fard S, Shamsi R, Seifi-Alan M, Javaheri M, Tabarestani S. Cancer-testis genes as candidates for immunotherapy in breast cancer. Immunotherapy. 2014;6(2):165–179. doi: 10.2217/imt.13.165. [DOI] [PubMed] [Google Scholar]
  • 33.Janosky M, Sabado RL, Cruz C, et al. MAGE-specific T cells detected directly ex-vivo correlate with complete remission in metastatic breast cancer patients after sequential immune-endocrine therapy. J. ImmunoTherapy Cancer. 2014;2(1):1–9. doi: 10.1186/s40425-014-0032-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fujita T, Yoshida A, Nishimura H, et al. Phase I clinical trial of multi-antigen peptide vaccines therapy using cancer-testis antigens for patients with advanced or recurrent breast cancer. J. Clin. Oncol. (Meeting Abstracts) 2012;30(15_suppl):e13037. [Google Scholar]
  • 35.US National Library of Medicine. MAGE-A in Breast Cancer. https://clinicaltrials.gov/ct2/results?term=MAGE&cond=Breast+Cancer
  • 36.US National Library of Medicine. NY-ESO-1 in Breast Cancer. https://clinicaltrials.gov/ct2/results?term=NY-ESO-1&cond=Breast+Cancer
  • 37.Liu W, Zhai M, Wu Z, et al. Identification of a novel HLA-A2-restricted cytotoxic T lymphocyte epitope from cancer-testis antigen PLAC1 in breast cancer. Amino Acids. 2012;42(6):2257–2265. doi: 10.1007/s00726-011-0966-3. [DOI] [PubMed] [Google Scholar]
  • 38.Neumann F, Kubuschok B, Ertan K, et al. A peptide epitope derived from the cancer testis antigen HOM-MEL-40/SSX2 capable of inducing CD4(+) and CD8(+) T-cell as well as B-cell responses. Cancer Immunol. Immunother. 2011;60(9):1333–1346. doi: 10.1007/s00262-011-1030-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sahin U, Tureci O, Chen YT, et al. Expression of multiple cancer/testis (CT) antigens in breast cancer and melanoma: basis for polyvalent CT vaccine strategies. Int. J. Cancer. 1998;78(3):387–389. doi: 10.1002/(SICI)1097-0215(19981029)78:3<387::AID-IJC22>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 40.Jager E, Chen YT, Drijfhout JW, et al. Simultaneous humoral and cellular immune response against cancer-testis antigen NY-ESO-1: definition of human histocompatibility leukocyte antigen (HLA)-A2-binding peptide epitopes. J. Exp. Med. 1998;187(2):265–270. doi: 10.1084/jem.187.2.265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Boel P, Wildmann C, Sensi ML, et al. BAGE: a new gene encoding an antigen recognized on human melanomas by cytolytic T lymphocytes. Immunity. 1995;2(2):167–175. doi: 10.1016/s1074-7613(95)80053-0. [DOI] [PubMed] [Google Scholar]
  • 42.Huang LQ, Brasseur F, Serrano A, et al. Cytolytic T lymphocytes recognize an antigen encoded by MAGE-A10 on a human melanoma. J. Immunol. 1999;162(11):6849–6854. [PubMed] [Google Scholar]
  • 43.Goodyear OC, Pearce H, Pratt G, Moss P. Dominant responses with conservation of T-cell receptor usage in the CD8+ T-cell recognition of a cancer testis antigen peptide presented through HLA-Cw7 in patients with multiple myeloma. Cancer Immunol. Immunother. 2011;60(12):1751–1761. doi: 10.1007/s00262-011-1070-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang L, Xu Y, Luo C, et al. MAGEA10 gene expression in non-small cell lung cancer and A549 cells, and the affinity of epitopes with the complex of HLA-A(*)0201 alleles. Cell. Immunol. 2015;297(1):10–18. doi: 10.1016/j.cellimm.2015.05.004. [DOI] [PubMed] [Google Scholar]
  • 45.Schuler MM, Nastke MD, Stevanovikc S. SYFPEITHI:database for searching and T-cell epitope prediction. Methods Mol. Biol. 2007;409:75–93. doi: 10.1007/978-1-60327-118-9_5. [DOI] [PubMed] [Google Scholar]
  • 46.Marcar L, Ihrig B, Hourihan J, et al. MAGE-A cancer/testis antigens inhibit MDM2 ubiquitylation function and promote increased levels of MDM4. PLoS One. 2015;10(5):e0127713. doi: 10.1371/journal.pone.0127713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Monte M, Simonatto M, Peche LY, et al. MAGE-A tumor antigens target p53 transactivation function through histone deacetylase recruitment and confer resistance to chemotherapeutic agents. Proc. Natl Acad. Sci. USA. 2006;103(30):11160–11165. doi: 10.1073/pnas.0510834103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hu W, Feng Z, Levine AJ. The regulation of multiple p53 stress responses is mediated through MDM2. Genes Cancer. 2012;3(3–4):199–208. doi: 10.1177/1947601912454734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Pellon-Maison M, Montanaro MA, Lacunza E, et al. Glycerol-3-phosphate acyltranferase-2 behaves as a cancer testis gene and promotes growth and tumorigenicity of the breast cancer MDA-MB-231 cell line. PLoS One. 2014;9(6):e100896. doi: 10.1371/journal.pone.0100896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Jagadish N, Gupta N, Agarwal S, et al. Sperm-associated antigen 9 (SPAG9) promotes the survival and tumor growth of triple-negative breast cancer cells. Tumour Biol. 2016;37(10):13101–13110. doi: 10.1007/s13277-016-5240-6. [DOI] [PubMed] [Google Scholar]
  • 51.Zhou L, Bao YL, Zhang Y, et al. Knockdown of TSP50 inhibits cell proliferation and induces apoptosis in P19 cells. IUBMB Life. 2010;62(11):825–832. doi: 10.1002/iub.390. [DOI] [PubMed] [Google Scholar]
  • 52.Shang B, Gao A, Pan Y, et al. 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]
  • 53.Xiao TZ, Bhatia N, Urrutia R, Lomberk GA, Simpson A, Longley BJ. MAGE I transcription factors regulate KAP1 and KRAB domain zinc finger transcription factor mediated gene repression. PLoS One. 2011;6(8):e23747. doi: 10.1371/journal.pone.0023747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yang B, O'herrin SM, Wu J, et al. MAGE-A, mMage-b, and MAGE-C proteins form complexes with KAP1 and suppress p53-dependent apoptosis in MAGE-positive cell lines. Cancer Res. 2007;67(20):9954–9962. doi: 10.1158/0008-5472.CAN-07-1478. [DOI] [PubMed] [Google Scholar]
  • 55.Song ZB, Ni JS, Wu P, et al. Testes-specific protease 50 promotes cell invasion and metastasis by increasing NF-kappaB-dependent matrix metalloproteinase-9 expression. Cell Death Dis. 2015;6:e1703. doi: 10.1038/cddis.2015.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Sugita Y, Wada H, Fujita S, et al. NY-ESO-1 expression and immunogenicity in malignant and benign breast tumors. Cancer Res. 2004;64(6):2199–2204. doi: 10.1158/0008-5472.can-03-3070. [DOI] [PubMed] [Google Scholar]
  • 57.Grigoriadis A, Caballero OL, Hoek KS, et al. CT-X antigen expression in human breast cancer. Proc. Natl Acad. Sci. USA. 2009;106(32):13493–13498. doi: 10.1073/pnas.0906840106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chen YT, Ross DS, Chiu R, et al. Multiple cancer/testis antigens are preferentially expressed in hormone-receptor negative and high-grade breast cancers. PLoS One. 2011;6(3):e17876. doi: 10.1371/journal.pone.0017876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lee HJ, Kim JY, Song IH, Park IA, Yu JH, Gong G. Expression of NY-ESO-1 in triple-negative breast cancer is associated with tumor-infiltrating lymphocytes and a good prognosis. Oncology. 2015;89(6):337–344. doi: 10.1159/000439535. [DOI] [PubMed] [Google Scholar]
  • 60.Curigliano G, Viale G, Ghioni M, et al. Cancer-testis antigen expression in triple-negative breast cancer. Ann. Oncol. 2011;22(1):98–103. doi: 10.1093/annonc/mdq325. [DOI] [PubMed] [Google Scholar]
  • 61.Curigliano G, Locatelli M, Fumagalli L, Goldhirsch A. Immunizing against breast cancer: a new swing for an old sword. Breast. 2009;18(Suppl. 3):S51–54. doi: 10.1016/S0960-9776(09)70273-5. [DOI] [PubMed] [Google Scholar]
  • 62.Yao J, Caballero OL, Yung WK, et al. Tumor subtype-specific cancer-testis antigens as potential biomarkers and immunotherapeutic targets for cancers. Cancer Immunol. Res. 2014;2(4):371–379. doi: 10.1158/2326-6066.CIR-13-0088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Balafoutas D, Zur Hausen A, Mayer S, et al. Cancer testis antigens and NY-BR-1 expression in primary breast cancer: prognostic and therapeutic implications. BMC Cancer. 2013;13:271. doi: 10.1186/1471-2407-13-271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.D'arcy V, Pore N, Docquier F, et al. BORIS, a paralogue of the transcription factor, CTCF, is aberrantly expressed in breast tumours. Br. J. Cancer. 2008;98(3):571–579. doi: 10.1038/sj.bjc.6604181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Walter S, Weinschenk T, Stenzl A, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat. Med. 2012;18(8):1254–1261. doi: 10.1038/nm.2883. [DOI] [PubMed] [Google Scholar]
  • 66.Wang Q, Jiang J, Ying G, et al. Tamoxifen enhances stemness and promotes metastasis of ERalpha36(+) breast cancer by upregulating ALDH1A1 in cancer cells. Cell Res. 2018 doi: 10.1038/cr.2018.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wang R, Yang L, Li S, et al. Quercetin inhibits breast cancer stem cells via downregulation of aldehyde dehydrogenase 1A1 (ALDH1A1), chemokine receptor type 4 (CXCR4), mucin 1 (MUC1), and epithelial cell adhesion molecule (EpCAM) Med. Sci. Monit. 2018;24:412–420. doi: 10.12659/MSM.908022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Croker AK, Rodriguez-Torres M, Xia Y, et al. Differential functional roles of ALDH1A1 and ALDH1A3 in mediating metastatic behavior and therapy resistance of human breast cancer cells. Int. J. Mol. Sci. 2017;18(10) doi: 10.3390/ijms18102039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Taghavi A, Akbari ME, Hashemi-Bahremani M, et al. Gene expression profiling of the 8q22–24 position in human breast cancer: TSPYL5, MTDH, ATAD2 and CCNE2 genes are implicated in oncogenesis, while WISP1 and EXT1 genes may predict a risk of metastasis. Oncol. Lett. 2016;12(5):3845–3855. doi: 10.3892/ol.2016.5218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Pugacheva EM, Teplyakov E, Wu Q, et al. The cancer-associated CTCFL/BORIS protein targets multiple classes of genomic repeats, with a distinct binding and functional preference for humanoid-specific SVA transposable elements. Epigenetics Chromatin. 2016;9(1):35. doi: 10.1186/s13072-016-0084-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tiffen JC, Bailey CG, Marshall AD, et al. The cancer-testis antigen BORIS phenocopies the tumor suppressor CTCF in normal and neoplastic cells. Int. J. Cancer. 2013;133(7):1603–1613. doi: 10.1002/ijc.28184. [DOI] [PubMed] [Google Scholar]
  • 72.Park S, Lim Y, Lee D, et al. Identification and characterization of a novel cancer/testis antigen gene CAGE-1. Biochim. Biophys. Acta. 2003;1625(2):173–182. doi: 10.1016/s0167-4781(02)00620-6. [DOI] [PubMed] [Google Scholar]
  • 73.Hitti E, Bakheet T, Al-Souhibani N, et al. Systematic analysis of AU-Rich element expression in cancer reveals common functional clusters regulated by key RNA-binding proteins. Cancer Res. 2016;76(14):4068–4080. doi: 10.1158/0008-5472.CAN-15-3110. [DOI] [PubMed] [Google Scholar]
  • 74.Zhou X, Yang F, Zhang T, et al. Heterogeneous expression of CT10, CT45 and GAGE7 antigens and their prognostic significance in human breast carcinoma. Jpn J. Clin. Oncol. 2013;43(3):243–250. doi: 10.1093/jjco/hys236. [DOI] [PubMed] [Google Scholar]
  • 75.Yamada R, Takahashi A, Torigoe T, et al. Preferential expression of cancer/testis genes in cancer stem-like cells: proposal of a novel sub-category, cancer/testis/stem gene. Tissue Antigens. 2013;81(6):428–434. doi: 10.1111/tan.12113. [DOI] [PubMed] [Google Scholar]
  • 76.Watkins J, Weekes D, Shah V, et al. Genomic complexity profiling reveals that HORMAD1 overexpression contributes to homologous recombination deficiency in triple-negative breast cancers. Cancer Discov. 2015;5(5):488–505. doi: 10.1158/2159-8290.CD-14-1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Khongkow P, Gomes AR, Gong C, et al. Paclitaxel targets FOXM1 to regulate KIF20A in mitotic catastrophe and breast cancer paclitaxel resistance. Oncogene. 2016;35(8):990–1002. doi: 10.1038/onc.2015.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brinkmann U, Vasmatzis G, Lee B, Pastan I. Novel genes in the PAGE and GAGE family of tumor antigens found by homology walking in the dbEST database. Cancer Res. 1999;59(7):1445–1448. [PubMed] [Google Scholar]
  • 79.Lu Y, Zhang K, Li C, et al. Piwil2 suppresses p53 by inducing phosphorylation of signal transducer and activator of transcription 3 in tumor cells. PLoS One. 2012;7(1):e30999. doi: 10.1371/journal.pone.0030999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Koslowski M, Sahin U, Mitnacht-Kraus R, Seitz G, Huber C, Tureci O. A placenta-specific gene ectopically activated in many human cancers is essentially involved in malignant cell processes. Cancer Res. 2007;67(19):9528–9534. doi: 10.1158/0008-5472.CAN-07-1350. [DOI] [PubMed] [Google Scholar]
  • 81.Bullinger L, Schlenk RF, Gotz M, et al. PRAME-induced inhibition of retinoic acid receptor signaling-mediated differentiation – a possible target for ATRA response in AML without t(15;17) Clin. Cancer Res. 2013;19(9):2562–2571. doi: 10.1158/1078-0432.CCR-11-2524. [DOI] [PubMed] [Google Scholar]
  • 82.Sinha A, Agarwal S, Parashar D, et al. Down regulation of SPAG9 reduces growth and invasive potential of triple-negative breast cancer cells: possible implications in targeted therapy. J. Exp. Clin. Cancer Res. 2013;32:69. doi: 10.1186/1756-9966-32-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Greve KB, Lindgreen JN, Terp MG, et al. Ectopic expression of cancer/testis antigen SSX2 induces DNA damage and promotes genomic instability. Mol. Oncol. 2015;9(2):437–449. doi: 10.1016/j.molonc.2014.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Xu H, Shan J, Jurukovski V, Yuan L, Li J, Tian K. TSP50 encodes a testis-specific protease and is negatively regulated by p53. Cancer Res. 2007;67(3):1239–1245. doi: 10.1158/0008-5472.CAN-06-3688. [DOI] [PubMed] [Google Scholar]

Articles from Immunotherapy are provided here courtesy of Taylor & Francis

RESOURCES