Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Oct 22.
Published in final edited form as: Nat Rev Urol. 2012 Jun 19;9(7):386–396. doi: 10.1038/nrurol.2012.117

Cancer/testis antigens and urological malignancies

Prakash Kulkarni 1, Takumi Shiraishi 2, Krithika Rajagopalan 3, Robert Kim 4, Steven M Mooney 5, Robert H Getzenberg 6
PMCID: PMC3477645  NIHMSID: NIHMS407159  PMID: 22710665

Abstract

Cancer/testis antigens (CTAs) are a group of tumour-associated antigens (TAAs) that display normal expression in the adult testis—an immune-privileged organ—but aberrant expression in several types of cancers, particularly in advanced cancers with stem cell-like characteristics. There has been an explosion in CTA-based research since CTAs were first identified in 1991 and MAGE-1 was shown to elicit an autologous cytotoxic T-lymphocyte (CTL) response in a patient with melanoma. The resulting data have not only highlighted a role for CTAs in tumorigenesis, but have also underscored the translational potential of these antigens for detecting and treating many types of cancers. Studies that have investigated the use of CTAs for the clinical management of urological malignancies indicate that these TAAs have potential roles as novel biomarkers, with increased specificity and sensitivity compared to those currently used in the clinic, and therapeutic targets for cancer immunotherapy. Increasing evidence supports the utilization of these promising tools for urological indications.

Introduction

As a result of the global increase in industrialization, urbanization, and acculturation changes in lifestyles, the worldwide incidence of cancer and the associated health-care costs are rising at an alarming rate. By 2030, the World Health Organization predicts that there will be more than 21 million new cancer cases and 13 million cancer-related deaths per year, generating a substantial cost burden to society.1 Furthermore, the combination of a rising global population and increased average life expectancy has led to, and continues to lead to, significant increases in the number of elderly patients with cancer, particularly those with urological cancers.2 According to a recent report commissioned by the National Institute on Aging (a division of the National Institute of Health in the USA), the number of people on this planet over 65 years of age will double to 1.4 billion (14% of the total population) in the next 30 years,3 underscoring the urgent need for new strategies to combat cancer.

Increased understanding of tumour genetics and pharmacological mechanisms of action, combined with technological advances in high-throughput genomics, have enhanced the development of new biomarkers and therapeutics. ‘Targeted therapies’ have been produced that are mainly, if not exclusively, active against tumours of a particular genotype, identified by a simple diagnostic test.4 In this era of ‘personalized’ medicine—which links individual patient biology with cancer risk, diagnosis, prognosis, and treatment—biomarkers play a critical role. Not only have biomarkers contributed greatly to our understanding of the heterogeneous nature of specific cancers, they have also led to significant improvements in treatment outcomes.5

Unfortunately, the biomarkers used to detect and treat urological malignancies—such as PSA and urine cytology—present several limitations relating to specificity, sensitivity, and cost-effectiveness. Furthermore, many of the therapeutic treatments currently in use— for example, androgen deprivation therapy (ADT) for prostate cancer68 and platinum compounds for bladder cancer9—are often discontinued as a result of drug resistance, following an initial period of positive response. Thus, biomarkers and therapeutic targets with increased efficacy and specificity—expressed highly in diseased tissue but not in surrounding normal tissue —are urgently needed.

A class of tumour-associated antigens (TAAs) called cancer/testis antigens (CTAs) has demonstrated potential in this context. These TAAs are typically expressed in embryonic stem cells and testicular germ cells, exhibiting little or no expression in most somatic tissues. They are also aberrantly expressed in several types of cancers.1014 Although cancer genes typically harbour mutations associated with their pathological function, mutations in genes encoding the CTAs are, to the best of our knowledge, extremely rare.1517 Nevertheless, CTAs are generally upregulated in response to DNA hypomethylation, which is a common feature of cancer cells.18,19 Indeed, treating cells with the DNA methyltransferase inhibitor decitabine (5-aza-2′-deoxycytidine) in vitro results in a dramatic upregulation of most, if not all, CTAs.20,21 Furthermore, several studies have demonstrated a correlation between CTA expression and promoter methylation status in tissue samples from patients with several different types of cancer.2224

Taken together, these observations suggest that the underlying cause of CTA-associated pathological function in cancer cells results from aberrant (dose-sensitive) expression of normal CTA genes, rather than the generation of mutated versions of these genes. In support of this theory, a study of fruit flies demonstrated a correlation between aberrant germline gene expression and tumorigenesis.25 Moreover, upregulated CTA expression has been associated with advanced disease and poorer prognosis,26 suggesting a key role of CTAs in tumorigenesis. Given the similarities between embryonic stem cells and cancer stem cells,2730 it seems likely that CTAs have an important function in disease progression and could, therefore, be useful for diagnosing and treating advanced cancers with stem-cell-like characteristics.

Broadly speaking, the CTAs can be grouped into two classes based on their chromosomal location. The CT-X antigens are located on the X chromosome and non-X CT antigens are located on the autosomes. Most CT-X antigens seem to be unique to primates and constitute several subfamilies of homologous genes, organized in discrete clusters along the X chromosome.31 As a result of their restricted expression in an immune-privileged organ, both groups of CTAs represent attractive immunotherapy targets.32 Antigenic CTA-derived peptides that are presented to the immune system with different human leukocyte antigen (HLA) allo-specificities elicit both humoral and cellular immune responses. Indeed, spontaneous humoral and cell-mediated immune responses have been demonstrated against several CTAs and patients with good antibody titres often present with better prognosis.33

A potential protocol to treat urological malignancies utilizes immunogenic peptides corresponding to CTAs (Figure 1). A tumour sample is first profiled for CTA expression using DNA microarrays and a CTA ‘finger-print’ is generated. Synthetic immunogenic peptides corresponding to the CTAs are then used to pulse in vitro immature dendritic cells isolated from the patient. The pulsed cells mature into professional antigen-presenting cells, which are then injected back into the patient to trigger an immune response. This strategy is applicable to most other nonurological cancers, provided specific CTAs are aberrantly expressed. At present, more than 200 CTAs have been described in the literature and two databases—CTpedia34 and ACTAbase35—serve as excellent resources on these antigens. Although emerging evidence has shed light on the functions of a few CT-X antigens in cancer, the majority remain poorly understood. By contrast, the non-X CTAs are fairly well conserved throughout evolution, with established roles in processes such as transformation,36 chromatin remodelling,32 transcriptional regulation, and signalling (Figure 2b).11 Thus, the non-X CT antigens are particularly promising targets for the development of small-molecule therapeutics.

Figure 1.

Figure 1

Schematic diagram of an immunotherapeutic approach to treating urological malignancies that utilizes immunogenic peptides corresponding to CTAs. A tumour sample is profiled for CTA expression using DNA microarrays. A CTA ‘fingerprint’ is generated and immunogenic peptides corresponding to the CTAs are generated that are used to pulse in vitro immature dendritic cells isolated from the patient. The pulsed cells mature into professional antigen presenting cells, which are then injected back into the patient to trigger an immune response. Abbreviations: CTA, cancer/testis antigen; Cy3, cyanine 3; Cy5, cyanine 5.

Figure 2.

Figure 2

Normalized expression heat maps. a | CT-X genes and b | non-X CT genes based on expression profiles from prostate cancer, bladder cancer, renal cell carcinoma, and testicular germ cell tumour samples (n = 397) obtained from the Gene Expression Omnibus (GEO) database.110 The complete list of expression profiles, together with the respective GEO dataset (GDS) records, is provided in the Supplementary Information online. The raw data files were converted into expression profiles via the GeneChip Robust Multi-Array Analysis method. Data analyses were performed using R packages,111 Bioconductor,112 and gplots.113 For each gene, the expression values from all samples were averaged for each type of urological malignancy. Mean expression levels for all genes were then normalized to range from 0 to 1. Some genes appear multiple times in this list as a result of different probes present on the chip.*Multiple genes involved.

Various CTA-based cancer vaccine trials are already underway (Table 1). Although several CTAs are differentially expressed in patients with urological malignancies (Figure 2), CTA-based therapy has been researched more extensively for other types of cancer—particularly melanoma—than for urological cancers. However, recent reports illustrate the significant progress that has been made with respect to the characterization of CTAs in patients with these malignancies. In this Review, we evaluate the available literature regarding the utility of CTAs as biomarkers and therapeutic tools for patients with prostate, bladder, renal cell, and testicular germ cell tumours (TGCTs).

Table 1.

Selected trials of cancer/testis antigen vaccines for various types of cancer

Vaccine Adjuvant Tumour histotype Phase Sponsor Trial identifier
GSK2132231A (MAGEA3 ASCI) Dacarbazine Melanoma I GSK NCT00849875
GSK1572932A (MAGEA3 ASCI) Cisplatin, vinorelbine and radiotherapy Non-small-cell lung cancer I GSK NCT00455572
CTAG1A-derived peptide or CTAG1A protein PF-3512676 and montanide ISA 720 VG (with or without cyclophosphamide) CTAG1A-expressing tumours I University of Pittsburgh NCT00819806
CTAG1A and CTAG2 peptides None Prostate cancer I Baylor College of Medicine NCT00711334
CTAG1A, gp100, and MART-1 peptides Oncovirus poly ICLC and montanide ISA 51 VG (with or without CP 870–893 antibody) Melanoma I H.3Lee Mof3tt Cancer Center and Research Institute NCT01008527
Recombinant MAGEA3 protein AS15 (with or without BCG) Bladder cancer I Patrice Jichlinski, University of Lausanne Hospitals NCT01498172
IMF-001 (recombinant CTAG1A protein and cholesteryl hydrophobized pullulan) None CTAG1A-positive solid tumours or melanoma I ImmunoFrontier, Inc. NCT01234012
CTAG1A genetically engineered T cells None Synovial carcinoma I University of Pennsylvania NCT01343043
Recombinant MAGEA3 protein AS15 Multiple myeloma I Ludwig Institute for Cancer Research NCT01380145
Mature dendritic cells pulsed with peptide mixes derived from full-length CTAG1A, MAGEA1, and MAGEA3 Decitabine (5-aza-2′-deoxycytidine) and imiquimod Neuroblastoma, Ewings sarcoma, osteogenic sarcoma, rhabdomyosarcoma, and synovial sarcoma I Penn State University NCT01241162
GSK2241658A (CTAG1A ASCI) None Melanoma I GSK NCT01213472
GSK2302032A (PRAME ASCI) None Non-small-cell lung cancer I GSK NCT01159964
Recombinant MAGEA3, MART1, and survivin Therapeutic autologous dendritic cells (with or without keyhole limpet haemocyanin) Melanoma I/II Dermatologische Klinik MIT Poliklinik, Universitaetsklinikum Erlangen NCT00074230
CTAG1A protein Poly-ICLC and montanide Melanoma I/II New York University Langone Medical Center NCT01079741
Autologous T3cells transduced with MAGEA3(A3A) and CTAG1A(C259) Cytoreductive chemotherapy Melanoma I/II University of Pennsylvania NCT01350401
PRS pan-DR, MAGEA3, MAGEA1, CTAG1A, and MART1 Cyclophosphamide Non-small-cell lung cancer II Institut Gustave Roussy NCT01159288
Recombinant MAGEA3 protein AS15 Urinary bladder neoplasms II European Association of Urology Research Foundation NCT01435356
GSK1203486A (MAGEA3 ASCI) None Melanoma II GSK NCT00706238
MAGEA3 and gp100 peptides Resiquimod (R848) Melanoma II M3D.3Anderson Cancer Center NCT00960752
MAGEA3 and CTAG1A peptides DT-PACE chemotherapy and autologous transplantation Multiple myeloma II/III University of Arkansas NCT00090493
GSK1572932A (MAGEA3 ASCI) None Non-small-cell lung cancer III GSK NCT00480025
GSK2132231A (MAGEA3 ASCI) None Melanoma III GSK NCT00796445

Abbreviations: ASCI, antigen-specific cancer immunotherapeutic; CTAG1A, cancer/testis antigen 1A; CTAG2, cancer/testis antigen 2; DT-PACE, dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide, and etoposide; gp100; glycoprotein 100; GSK, Glaxo Smith Kline; MAGE, melanoma antigen family proteins; MART1, melanoma antigen recognized by T cells 1; PRAME, preferentially expressed antigen in melanoma; PRS pan-DR; proline-rich sequence pan-DR.

CTAs and prostate cancer

Worldwide, prostate cancer is the most common urological malignancy. In men, it is the second most frequently diagnosed cancer and the sixth leading cause of cancer death, with 903,500 new cases and 258,400 cancer deaths reported in 2008.2 The introduction of serum PSA testing is thought by many to have dramatically improved the prognosis of men diagnosed with prostate cancer. However, two recent reports from Europe and the USA have sparked controversy regarding this issue.32,33 Although the European study concluded that routine PSA screening reduced the mortality rate associated with prostate cancer by 20%,37 the US-based population study showed no discernable difference between patients who received PSA screening and control groups.38 Consistent with the latter findings, the US Preventive Services Task Force (USPSTF) has now officially recommended against routine PSA screening for prostate cancer. Although this recommendation remains equivocal, it is generally accepted that the widespread use of PSA testing results in the over detection and over treatment of potentially indolent disease. On the other hand, the fact that relatively few men present with metastatic disease has been attributed, at least in part, to PSA screening protocols.39

ADT is the gold-standard first-line treatment for metastatic prostate cancer. Although this approach is usually effective to begin with, the response to ADT is often transient and tumours tend to progress to castration-resistant prostate cancer (CRPC). Despite improvements to chemotherapy regimens, CRPC is characteristically life threatening.6,7 Thus, there are two major challenges facing the management of prostate cancer today; firstly, to develop new biomarkers that can identify aggressive phenotypes of prostate cancer and, secondly, to develop new treatment options for CRPC.

CTAs as biomarkers

Several studies have investigated the potential role of CTAs as biomarkers in prostate cancer. In early reports, protein expression of cancer/testis antigen 1A (CTAG1A) was detected in 3% of patients with localized prostate cancer and 15% of patients with CRPC.40 Subsequently, Hudolin et al.41 reported that, although expression of melanoma antigen family A, 1 (MAGEA1) was observed for just 10.8% of carcinoma samples, antibodies against multiple MAGEA antigens and combinations of CTAG1A and cancer testis antigen 2 (CTAG2) stained 85.9% and 84.8% of samples, respectively. Synovial sarcoma X protein expression has been shown to be largely restricted to metastatic prostate cancers, further supporting a role for CTAs as biomarkers of disease progression.42

In order to elucidate CTA expression patterns in prostate cancer samples, Suyama et al.24 produced a customized DNA microarray tiled with DNA probes representing a significant portion of all known CTAs. This study revealed that several CTAs are upregulated in prostate cancer cells, the majority of which are CT-X antigens. Several of the MAGEA and chondrosarcoma-associated gene subfamilies of CT-X antigens were upregulated in CRPCs but not in primary prostate cancers, whereas P antigen family, member 4 (prostate associated; PAGE4) expression was, conversely, upregulated in primary prostate cancers but silent in metastatic disease. These observations suggest that CTAs could potentially be used as biomarkers to distinguish men with aggressive disease (who are likely to require treatment) from men with more indolent disease (for whom a conservative treatment programme, such as active surveillance, might be more appropriate).

To further investigate this potential role, Shiraishi et al.43 used a panel of five CTAs—centrosomal protein 55 kDa (CEP55), NUF2, NDC80 kinetochore complex component, homolog (S. cerevisiae; NUF2), PAGE4, PDZ binding kinase (PBK) and TTK protein kinase (TTK)—that are differentially expressed in prostate cancer to devise a multiplex real-time PCR assay to predict prostate cancer progression and identify disease recurrence following radical prostatectomy. Expression of all of the CTAs, with the exception of TTK, significantly correlated with prostatectomy Gleason score using Pearson’s correlation coefficient (CEP55 P = 0.0057; NUF2 P = 0.0051; PBK P = 0.0295; PAGE4 P = 0.0236). However, CTA expression was not shown to correlate with age, preoperative serum PSA level, or tumour stage. Although Gleason score is thought to be one of the strongest predictors of recurrence, it tends to be relatively subjective. The multiplex assay could provide an objective assessment that is quantitative, consistent across institutions, and perhaps even useful as a molecular surrogate for Gleason score. However, it is worth noting that the investigators reported several limitations to the study, including a small sample size and a sample set that was nonreflective of a contemporary series.43,41 Additional studies—performed across multiple institutions and involving larger contemporary-matched cohorts of patients presenting with newly diagnosed prostate cancer—could further validate this CTA-based predictive test.

Using a different approach, Xie et al.44 developed a multiplex assay that simultaneously measures expression of autoantibodies against CTAG1A, X antigen family, member 1B (XAGE1B), synovial sarcoma, X breakpoint 2 (SSX2), synovial sarcoma, X breakpoint 4 (SSX4), alpha-methylacyl-CoA racemase (AMACR), p90 autoantigen, PC4 and SFRS1 interacting protein 1 (PSIP1; also known as LEDGF) and PSA. This assay demonstrated enhanced sensitivity and specificity over PSA alone with respect to distinguishing prostate cancer from nonmalignant tissue. The presence of a natural immune response against CTAs could, therefore, represent an alternative strategy for exploiting these antigens as diagnostic and prognostic markers for prostate cancer.

Despite these promising preliminary findings, several limitations to the use of CTAs as biomarkers for prostate cancer have already been identified. For example, the frequency of CTA expression is not generally high enough for individual CTAs to serve as single biomarkers; a problem that is exacerbated by the heterogeneity of prostate cancer. Thus, combining the expression analysis of a CTA with that of other CTAs (or non-CTA markers) might be necessary to exploit CTAs as biomarkers for prostate cancer.

CTAs as therapeutic targets

The efficacy of sipuleucel-T—an autologous cellular immunotherapy—for the treatment of men with asymptomatic or minimally symptomatic metastatic CRPC has set a precedence for prostate cancer immunotherapy. This treatment approach is emerging as a promising modality for chemotherapy-resistant prostate cancer and perhaps even early-stage disease. Accumulating evidence suggests that CTAs could be the next generation of immunotherapeutic targets for advanced prostate cancer. CTAG1A and members of the MAGE family have already been extensively studied as therapeutic targets for other types of cancer in several clinical trials4547 and CTAG1A has recently entered a phase I clinical trial for prostate cancer (Table 1).48

In other studies, aimed at detecting circulating auto-antibodies, anti-CTAG1A antibodies have been observed more frequently in the serum of patients with advanced disease (18.9%) compared to localized tumours (0.9%).40 Similarly, SSX2 antibodies and SSX2-specific T cells have been identified in the peripheral blood of patients with prostate cancer, indicating that patients can have pre- existing immunity to SSX2.42,49,50 T-cell-mediated immune responses to PAGE4 have also been reported in patients with prostate cancer.51 Parmigiani et al.52 described a novel CTA, ankyrin repeat domain 30B pseudogene 2 (ANKRD30BP2; also known as CTSP-1), which is aberrantly expressed in 58% of prostate tumors and capable of eliciting a humoral immune response (HIR) in about 20% of patients with prostate cancer. The same group also reported that patients with anti-CTSP-1 antibodies have a lower risk of biochemical recurrence and that presence of anti-CTSP-1 antibodies significantly associated with a better prognosis in patients with higher Gleason grade tumours.53

Furthermore, centrosomal protein 290 kDa (CEP290; also known as T21) was identified by the SEREX (Serological Analysis of Recombinant cDNA EXpression libraries) approach, which can identify tumour antigens that can elicit high titre IgG antibody responses by combining serological analysis with antigen cloning techniques. 54 In this study, which utilized a normal testicular cDNA library and pooled allogeneic sera from patients with prostate cancer, CEP290 was shown to be significantly overexpressed in patients with prostate cancer compared to those with benign prostate epithelium. An HIR against CEP290 was observed in 50% of patients with prostate cancer. Moreover, one research team detected A kinase (PRKA) anchor protein 4 (AKAP4)— which induces AKAP4-specific and major histocompatibility complex, class I, A (HLA-A)-restricted cytotoxic T lymphocytes (CTLs)—in 65% of prostate cancer tissues (13 of 20).55 Taken together, these findings suggest that several CTAs are potential immunotherapeutic candidates that await clinical validation for advanced prostate cancer.

In a phase I clinical trial, 24 patients with metastatic CRPC were treated with ipilimumab—an antibody that blocks CTL-associated antigen 4 (CTLA4)—and granulocyte–macrophage colony-stimulating factor (GM-CSF).56 PSA declines of more than 50% were observed in 50% of patients treated with the highest doses (n = 6). Interestingly, IgG antibodies against CTAG1A were induced in one of the three clinical responders. These results show that this combination immunotherapy with CTLA4 blockade and GM-CSF can induce the expansion, not only of activated effector CD8+ T cells, but also of CTA-specific T cells. Although the physiological functions of most CTAs—especially the CT-X antigens—remain poorly understood,11,26 data are beginning to shed new light on immunotherapy approaches for prostate cancer. For example, among the CT-X antigens, MAGEA11 was shown to increase androgen receptor (AR) transcriptional activity via direct interactions with AR and other coactivators.5759 Suyama et al.24 demonstrated that MAGEA2 is associated with proliferation and chemosensitivity of prostate cancer cells. Similarly, expression of members of the SSX protein family has been associated with stem cell migration, suggesting a biologically important role that is connected to its metastatic phenotype.60 Among the non-X CT antigens, lysine (K)-specific demethylase 5B (KDM5B; also known as JARID1B)—a chromatin remodeller that specifically demethylates histone H3 lysine 4 (H3K4)— is upregulated in prostate cancers (especially metastatic tumours) and potentiates AR transactivation.61 Collectively, these observations underscore the potential for discovering small molecule therapeutics for prostate cancer by targeting the CTAs.

CTAs and bladder cancer

Bladder cancer, which is reported more frequently in men than in women, is the second most common urological malignancy (after prostate cancer). In 2008, 386,300 new cases of bladder cancer and 150,200 associated deaths were reported worldwide.2 Three main types of bladder cancer have been described; transitional cell carcinoma (TCC), squamous cell carcinoma, and adenocarcinoma. TCC, which accounts for about 90% of bladder cancer cases, mainly occurs in the innermost lining of the bladder and can be either superficial (70–80% of cases), muscle invasive, or metastatic. Adenocarcinoma, which accounts for less than 2% of cases, arises from glandular cells.62 Although several biomarkers have been identified for bladder cancer, no single biomarker is a reliable predictor of disease recurrence or prognosis, and false-negative rates are generally high. The pathologic diagnosis achieved with cystoscopy still remains the gold standard for detecting and evaluating both early and late stages of bladder cancer. However, although highly specific (90% specificity), this approach is associated with poor sensitivity and is an invasive technique that requires the insertion of a catheter.63

CTAs as biomarkers

Depending on the type and stage of the bladder tumour, different treatment options are administered. Superficial papillary TCC is often treated with transurethral resection of bladder tumour (TURBT).63 Superficial non-papillary TCC—which is generally more aggressive and tends to progress to invasive tumours—is often treated with BCG, a form of immunotherapy.64 In some patients, BCG treatment has proven to be helpful in reducing the risk of recurrence and progression after TURBT.65 Invasive TCCs are often treated with radical cystectomy in combination with radiation and chemotherapy. Although initial response to chemotherapeutic agents (including platinum compounds) is typically good, treatment does not generally lead to a cure and tumour recurrence is typical.9 Hence, biomarkers with high sensitivity and specificity are urgently needed. It is also important to be able to differentiate between patients with invasive and superficial TCCs (to avoid overtreating or undertreating patients), identify patients who are likely to respond to immunotherapy (to reduce unnecessary adverse effects), and to determine the most appropriate timing for radical cystectomy.

Several CTAs, especially CT-X antigens, are highly expressed in bladder cancers (Figure 2).26 One early study reported increased expression of MAGEA1 (21% of cases) and MAGEA3 (35% of cases) in primary TCCs.66 Subsequently, increased expression of several MAGE genes has been associated with advanced stages of the disease.67 In these immunohistochemical studies, several members of the MAGE gene family were highly expressed in squamous cell carcinoma and TCC compared with normal bladder tissue. Of particular note was MAGEA9, which was highly expressed in the majority of patients (>60%) with higher grades (2 and 3) of TCC and associated lymph node metastases. Increased expression of both MAGEA9 and MAGEA4 was associated with progression to metastatic disease, as well as disease recurrence. Thus, MAGEA9 could be useful, not only as a prognostic and diagnostic marker, but also as an immunotherapeutic target.68

Another member of the MAGEA family, MAGEA10, seems to be the most immunogenic of the MAGE antigens and is upregulated in subsets of both noninvasive (19.4%) and invasive (31.3%) bladder cancers compared to normal bladder tissue.69 In this study, MAGEA10 expression was used to measure the effectiveness of immunotherapy, underscoring its biomarker potential. Another CT-X antigen that is frequently overexpressed in a variety of tumours is CTAG1A.70 This CTA was shown to demonstrate higher expression in TCC than in squamous cell carcinoma using epitope mapping and tissue microarray analysis.71 Some of the non-X CT antigens have also been shown to be highly expressed in bladder cancers. For example, acrosin binding protein (ACRBP) is overexpressed in 28% of bladder cancers,72 and lymphocyte antigen 6 complex, locus K (LY6K), a putative oncogene, is highly upregulated in clinical samples from patients with bladder tumours, as well as in several bladder cancer cell lines.73 JARID1B was shown to be significantly upregulated in bladder cancer specimens compared to normal tissue, with a dose-sensitive effect in several bladder cancer cell lines.74

CTAs as therapeutic targets

On the basis that MAGE genes are highly expressed in advanced tumours, Nishiyama et al.75 used autologous dendritic cells pulsed with MAGEA3 peptide as an immunotherapeutic option. The investigators established that, in 75% of treated patients (n = 4), lymph node metastases and liver metastases were significantly reduced, with no severe adverse effects.75 As noted earlier, expression levels of CTAs are often upregulated in response to epigenetic modulators. Picard et al.76 demonstrated that MAGEA9 expression was increased in response to the DNA methyltransferase inhibitor decitabine and histone deacetylase inhibitors MS-275 and 4-phenylbutyrate, highlighting their potential for the targeted treatment of bladder cancer.

CTAs and renal cell carcinoma

Kidney cancer accounts for about 2% of all new cancer cases worldwide,77 with 111,100 cases and 43,000 deaths reported in 2008 in the developed world.2 Renal cell carcinoma (RCC) accounts for approximately 85% of all kidney cancers; the remaining 15% includes transitional cell cancers of the renal pelvis (approximately 12%), sarcomas, and other soft tissue malignancies (approximately 2%).78 At present, surgical resection is the most effective treatment for localized RCC tumours; 5-year survival is 85% for patients with organ-confined disease who undergo nephrectomy.79,80 However, one-third of patients have metastases at diagnosis and, among those with clinically localized disease, 30–40% will develop metastases after surgery.76,81 Treatment options in these patients remain limited and 5-year survival is estimated at less than 5%.82 Furthermore, RCC is resistant to conventional therapies such as radiation and chemotherapy83,84 and prognosis for patients with advanced stages of RCC remains poor, underscoring the urgent need for novel diagnostic and prognostic biomarkers and therapeutic targets.

CTAs as biomarkers

CTA expression profiles vary with cancer type, enabling the classification of cancers based on the frequency of CTA expression. On this basis, RCC is characterized as a cancer type with low expression of relatively few CTAs; thus, the utility of CTAs as biomarkers for RCC seems to be limited.26 Consistent with this prediction, a study involving 57B—a murine pan-specific monoclonal antibody that has been shown to react with MAGEA1, MAGEA3, MAGEA4, MAGEA6, and MAGEA12— demonstrated that none of these CT-X antigens are expressed in RCCs.67 Furthermore, the same group also used IHC analysis to show that CTAG1A expression is not upregulated in RCCs.85

However, in contrast to these findings, Yamanaka et al.86 demonstrated that MAGEA1, MAGEA2, MAGEA3, and MAGEA4 genes were expressed in 22%, 16%, 76%, and 30% of RCC samples tested using reverse transcription PCR (n = 50) and Garg et al.87 reported expression of sperm associated antigen 9 (SPAG9) in 88% of patients with RCC using in situ RNA hybridization and immunohistochemistry. An HIR against SPAG9 was also detected in the sera of patients with RCC—but not in healthy individuals—by enzyme-linked immunosorbent assay and immunoblotting. Furthermore, recent work from our laboratory has shown that the non-X CT antigen SPAG4 is highly expressed in RCC and regulated by hypoxia inducible factor. Interestingly, low levels of SPAG4 mRNA expression significantly correlate with shorter progression free survival, suggesting that SPAG4 could serve as a promising diagnostic and prognostic biomarker for RCC (P. Kulkarni, unpublished work). These conflicting data can be explained by differences in the sensitivities of the techniques employed, emphasizing the need for additional studies to evaluate the potential of CTAs as biomarkers for RCC.

CTAs as therapeutic targets

Given that RCC represents one of the most immuneresponsive cancers, immunotherapy is a promising strategy for treating patients with RCC.88,89 However, as a result of low expression frequencies, the use of CTAs as immunotherapy targets presents additional challenges. Nonetheless, treatment with the decitabine has been shown to induce CTAG1A expression for up to 60 days following treatment, generating a functional protein that is efficiently recognized by HLA-A2-restricted CTAG1A-specific CTLs. This finding supports the use of CTAs as therapeutic targets for RCC.90

CTAs and testicular germ cell tumours

Testicular cancer is the most prevalent cancer in men aged 15–44 years, with an average of 4.6 cases and 0.3 deaths per 100,000 men reported in 2008.2,91,92 Treatment is successful in virtually 100% of patients with organ-confined disease and 80% of those with metastatic disease.93 However, there are two major challenges that face testicular cancer care; identifying patients that can be treated with orchiectomy alone (without the need for chemotherapy or radiation) and, if disease is metastatic, determining which patients have responded well to nonsurgical treatment (radiation or chemotherapy) and do not require invasive surgery to remove metastases.

Both major forms of TGCTs—nonseminomas and seminomas—are generally diagnosed at a relatively young age (median age of 25 and 35 years, respectively), whereas spermatocytic seminomas—a much less common form of testicular cancer—affects men aged more than 50 years.94 Nonseminomas and seminomas often metastasize and are generally treated by orchiectomy and platinum-based therapies. These malignancies are thought to arise from primitive gonocytes and primordial germ cells (PGCs), which are present before birth (Figure 3). This suggests that intratubular germ cell neoplasia unclassified cells, which give rise to seminomas and nonseminomas but not spermatocytic seminomas, actually form during development and are thus termed inaccurately. Spermatocytic seminomas resemble adult spermatogonia and spermatocytes, and probably develop from these cells much later in life than other testicular malignancies. It is, therefore, not surprising that non-seminomas and seminomas are the most aggressive TGCTs, followed by spermatocytic seminomas.94 In fact, spermatocytic seminomas most frequently present in older men as painless enlargements of the testis that rarely metastasize and are, as such, generally treated with orchiectomy alone.

Figure 3.

Figure 3

Schematic diagram of mammalian spermatogenesis that depicts the stages of testicular germ cell tumour development. Primordial germ cells enter the testis during embryogenesis and develop into gonocytes. These cells, which are only present before birth, are thought to be the precursors of seminomas and nonseminomas. In humans, the process of spermatogenesis begins at puberty, from precursor cells known as spermatogonia, and continues throughout the adult life. Spermatocytic seminoma is thought to develop from spermatogonia or spermatoytes.

CTAs as biomarkers

The use of biomarkers for detecting and monitoring TGCTs has been limited to date. TGCT screening is largely restricted to periodic digital self-examination, as recommended by general practitioners. In order to confirm diagnosis and prognosis after surgery, a blood test for α-fetoprotein and β-human chorionic gonadotropin is commonly performed following treatment (orchiectomy with chemotherapy or radiation). However, these biomarkers are rarely seen in seminomas and are not necessarily expressed in nonseminomas.95

Although CTAs are not currently used as markers for TGCT, some studies have produced fairly promising data to support their use—especially for seminomas. As outlined in Supplementary Table 1 online, these tumours normally express cancer/testis antigen family 45 (CT45) members, G antigen (GAGE) family members, MAGEB1, MAGEB2, MAGEC1, MAGEC2, and testis specific protein, Y-linked (TSPY) family members. Although these CTAs seem to be excellent tissue markers, their use as serum-based markers has not yet been investigated. Several laboratories have also demonstrated that CTA expression coincides with the repression of pluripotency genes during sperm development and similarly, during tumorigenesis of their respective cancer lineages.94 OCT3/4 and NANOG— transcription factors that trigger the development of induced pluripotent stem (iPS) cells—are upregulated in many TGCTs (including seminomas and nonseminomas, but not spermatocytic seminomas).7,96,97 It is not surprising then that spermatocytic seminomas actually express a wider array of CTAs than seminomas and nonseminomas (Supplementary Table 1 online). Increased transition from pluripotency to CTA expression in spermatocytic seminomas could explain the improved prognosis observed in patients who undergo orchiectomy alone compared to seminomas and nonseminomas, which also require adjuvant therapy.

CTAs as therapeutic targets

Given the immune-privileged nature of the testis, CTAs are potential therapeutic targets for non-organ-confined disease. However, as metastatic disease is already highly sensitive to platinum-based therapies and radiation, only a subset of metastatic cancers (about 20%) will require further treatment.95 These cases, which are recurrent and generally fatal, might be treatable by CTA-based immunotherapy.

Conclusions

Therapeutic vaccination for cancer treatment depends on T-cell response to TAAs.98 Thus, the identification and characterization of immunogenic CTA peptides, together with the identification of respective HLA class I antigen restriction, is a highly viable approach for cancer immunotherapy (Figure 1). Unfortunately, several issues have hampered the success of this approach in a clinical setting. Firstly, low levels of expression, coupled with great intertumoural and intratumoural heterogeneity in CTA expression, mean that these approaches are some-what challenging. Secondly, CTA expression heterogeneity could also impair immunogenicity and immune recognition of cancer cells by the immune system, resulting in decreased vaccination efficacy. An even bigger potential problem is that tumour cell heterogeneity could lead to the emergence of CTA-negative neoplastic clones, capable of escaping treatment-induced and CTA-specific immune surveillance.99

A possible solution to these limitations is to boost CTA expression by administering decitabine, a DNA hypomethylating agent used to treat myelodysplastic syndromes.100 In addition to their effect on CTA expression, such agents are characterized by a more general immunomodulatory response, which leads to the persistent upregulation of molecules involved in tumour antigen presentation.101 Thus, the ability to pharmacologically ‘normalize’ intratumoural CTA distribution and upregulate the expression of HLA antigens, accessory molecules, and costimulatory molecules has fuelled optimism for the use of CTA-based vaccination therapy.99 Furthermore, the use of these epigenetic modulators is likely to render cancer cells more immunogenic and less prone to immune surveillance evasion. However, such optimism needs to be balanced with caution. For example, there could be ‘off-target’ effects of epigenetic drugs that could, potentially, result in undesirable consequences. Additionally, some CTAs have putative oncogenic functions when overexpressed, even if only transiently, which could be counterproductive. The benefits and risks of such an approach must, therefore, be carefully contemplated.

Tumour cell heterogeneity is also a major impediment to developing reliable biomarkers. Using a combination of markers is likely to improve the accuracy of prognosis and treatment stratification compared to utilization of a single marker. In fact, there has already been some success in the identification of subtypes of certain urological malignancies, such as prostate cancer, based on genetic signatures.102,103 In the future, CTA-based gene signatures could be used to complement clinicopathological factors in the diagnosis of aggressive and indolent cancers, discerning treatment responders from nonresponders, and monitoring therapeutic response. Perhaps more robust mRNA-based platforms designed for high-throughput screening with greater sensitivity than typical DNA microarrays104—such as the nCounter® Gene Expression Assay (NanoString Technologies, Seattle, WA, USA)—could be utilized to achieve highly reproducible results across multiple institutions. The efficacy of such platforms could be tested directly using total blood samples105,106 or circulating tumour cells,107 providing critical information regarding diagnosis and prognosis in a minimally invasive manner.

The lack of knowledge regarding the functions of most CTAs, especially in relation to the development of cancer, has limited progress in targeting them therapeutically. However, work from our laboratory has identified some interesting characteristics of CTAs that shed new light on their functions. For example, more than 90% of CTAs are predicted to be intrinsically disordered proteins (IDPs).108 IDPs are proteins that lack a rigid structure, at least in vitro, and are frequently overexpressed in several pathological conditions. The inherent ability of IDPs to engage in promiscuous interactions with other proteins when present at high concentrations is thought to explain their pathological effects.109 Taken together, these observations provide a novel perspective on CTAs, implicating them in the dose-sensitive processing and transducing of information in cells with altered physiological states. These findings should enhance our understanding of the functions of CTAs and facilitate the development of novel cancer therapeutics.

Key points.

  • Restricted expression of cancer/testis antigens (CTAs) in an immune-privileged organ (the adult testis) and aberrant expression in malignant tissue make CTAs ideal biomarkers and immunotherapeutic targets for managing urological cancers

  • Unlike many cancer-associated genes, which frequently harbour mutations associated with their pathological function, mutations in genes encoding the CTAs are extremely rare

  • The majority of CTAs are thought to be intrinsically disordered proteins that often engage in promiscuous interactions with other proteins when overexpressed

  • A key problem for CTA-based therapies is tumour cell heterogeneity, which results in differential expression of CTAs; use of the FDA-approved DNA methylation inhibitor decitabine can circumvent this issue

  • Use of a combination of markers is likely to improve the accuracy of prognostication and treatment stratification compared to utilization of a single marker

  • The success of clinical trials underscores the immunotherapeutic potential of a CTA-based approach and indicates that CTAs could be used as clinical tools for urological malignancies in the near future

Review criteria.

We conducted detailed searches of the medical literature published in PubMed up until March 31st 2012. The search terms we used were “cancer/testis antigens”, “urological malignancies”, “prostate cancer”, “bladder cancer”, “testicular cancer”, “renal cell carcinoma”, “kidney cancer”, “biomarker”, and “therapeutic target”. Only English-language articles were included.

Acknowledgments

This work was supported by a National Cancer Institute Specialized Program of Research Excellence, the Bernard L. Schwartz Scholar Award by the Patrick C. Walsh Cancer Research Fund, and the Patana Fund of the Brady Urological Institute.

Footnotes

Competing interests

P. Kulkarni, T. Shiraishi and R. H. Getzenberg declare associations with the following organization: Johns Hopkins University. See the article online for full details of the relationships.

Author contributions

All authors contributed equally to researching the article and discussions of content, as well as the writing and editing of the manuscript prior to submission.

Supplementary information

Supplementary information is linked to the online version of the paper at www.nature.com/reviews/nrurol.

Contributor Information

Prakash Kulkarni, James Buchanan Brady Urological Institute, 600 North Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.

Takumi Shiraishi, James Buchanan Brady Urological Institute, 600 North Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.

Krithika Rajagopalan, James Buchanan Brady Urological Institute, 600 North Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.

Robert Kim, Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.

Steven M. Mooney, James Buchanan Brady Urological Institute, 600 North Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA

Robert H. Getzenberg, James Buchanan Brady Urological Institute, 600 North Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA

References

  • 1.O’Callaghan T. Introduction: the prevention agenda. Nature. 2011;471:2–4. doi: 10.1038/471S2a. [DOI] [PubMed] [Google Scholar]
  • 2.Jemal A, et al. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
  • 3.Kinsella K, Wan HE. International population report. An aging world. U.S. Government Printing Office; Washington DC: 2009. [Google Scholar]
  • 4.Arteaga CL, Baselga J. Impact of genomics on personalized cancer medicine. Clin Cancer Res. 2012;18:612–618. doi: 10.1158/1078-0432.CCR-11-2019. [DOI] [PubMed] [Google Scholar]
  • 5.Parkinson DR, Johnson BE, Sledge GW. Making personalized cancer medicine a reality: challenges and opportunities in the development of biomarkers and companion diagnostics. Clin Cancer Res. 2012;18:619–624. doi: 10.1158/1078-0432.CCR-11-2017. [DOI] [PubMed] [Google Scholar]
  • 6.Tannock IF, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351:1502–1512. doi: 10.1056/NEJMoa040720. [DOI] [PubMed] [Google Scholar]
  • 7.Aragon-Ching JB, Dahut WL. Chemotherapy in androgen-independent prostate cancer (AIPC): what’s next after taxane progression? Cancer Ther. 2007;5:151–160. [PMC free article] [PubMed] [Google Scholar]
  • 8.Foley R, Marignol L, Keane JP, Lynch TH, Hollywood D. Androgen hypersensitivity in prostate cancer: molecular perspectives on androgen deprivation therapy strategies. Prostate. 2011;71:550–557. doi: 10.1002/pros.21266. [DOI] [PubMed] [Google Scholar]
  • 9.Zachos I, et al. Systemic therapy of metastatic bladder cancer in the molecular era: current status and future promise. Expert Opin Investig Drugs. 2010;19:875–887. doi: 10.1517/13543784.2010.496450. [DOI] [PubMed] [Google Scholar]
  • 10.Scanlan MJ, et al. Identification of cancer/testis genes by database mining and mRNA expression analysis. Int J Cancer. 2002;98:485–492. doi: 10.1002/ijc.10276. [DOI] [PubMed] [Google Scholar]
  • 11.Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer. 2005;5:615–625. doi: 10.1038/nrc1669. [DOI] [PubMed] [Google Scholar]
  • 12.Bera TK, et al. Selective POTE paralogs on chromosome 2 are expressed in human embryonic stem cells. Stem Cells Dev. 2008;17:325–332. doi: 10.1089/scd.2007.0079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gjerstorff MF, et al. Distinct GAGE and MAGE-A expression during early human development indicate specific roles in lineage differentiation. Hum Reprod. 2008;23:2194–2201. doi: 10.1093/humrep/den262. [DOI] [PubMed] [Google Scholar]
  • 14.Lifantseva N, et al. Expression patterns of cancer-testis antigens in human embryonic stem cells and their cell derivatives indicate lineage tracks. Stem Cells Int. doi: 10.4061/2011/795239. http://dx.doi:10.4061/2011/795239. [DOI] [PMC free article] [PubMed]
  • 15.Caballero OL, et al. Frequent MAGE mutations in human melanoma. PLoS ONE. 2010;5:e12773. doi: 10.1371/journal.pone.0012773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Clark J, et al. Identification of novel genes, SYT and SSX, involved in the t(X;18)(p11.2;q11.2) translocation found in human synovial sarcoma. Nat Genet. 1994;7:502–508. doi: 10.1038/ng0894-502. [DOI] [PubMed] [Google Scholar]
  • 17.Cerveira N, et al. A novel spliced fusion of MLL with CT45A2 in a pediatric biphenotypic acute leukemia. BMC Cancer. 2010;10:518. doi: 10.1186/1471-2407-10-518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sigalotti L, et al. Promoter methylation controls the expression of MAGE2, 3 and 4 genes in human cutaneous melanoma. J Immunother. 2002;25:16–26. doi: 10.1097/00002371-200201000-00002. [DOI] [PubMed] [Google Scholar]
  • 19.Sigalotti L, et al. Intratumor heterogeneity of cancer/testis antigens expression in human cutaneous melanoma is methylation-regulated and functionally reverted by 5-aza-2′-deoxycytidine. Cancer Res. 2004;64:9167–9171. doi: 10.1158/0008-5472.CAN-04-1442. [DOI] [PubMed] [Google Scholar]
  • 20.Karpf AR. A potential role for epigenetic modulatory drugs in the enhancement of cancer/germ-line antigen vaccine efficacy. Epigenetics. 2006;1:116–120. doi: 10.4161/epi.1.3.2988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Akers SN, Odunsi K, Karpf AR. Regulation of cancer germline antigen gene expression: implications for cancer immunotherapy. Future Oncol. 2010;6:717–732. doi: 10.2217/fon.10.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yegnasubramanian S, et al. DNA hypomethylation arises later in prostate cancer progression than CpG island hypermethylation and contributes to metastatic tumor heterogeneity. Cancer Res. 2008;68:8954–8967. doi: 10.1158/0008-5472.CAN-07-6088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Glazer CA, et al. Integrative discovery of epigenetically derepressed cancer testis antigens in NSCLC. PLoS ONE. 2009;4:e8189. doi: 10.1371/journal.pone.0008189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Suyama T, et al. Expression of cancer/testis antigens in prostate cancer is associated with disease progression. Prostate. 2010;70:1778–1787. doi: 10.1002/pros.21214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Janic A, Mendizabal L, Llamazares S, Rossell D, Gonzalez C. Ectopic expression of germline genes drives malignant brain tumor growth in Drosophila. Science. 2010;330:1824–1827. doi: 10.1126/science.1195481. [DOI] [PubMed] [Google Scholar]
  • 26.Scanlan MJ, Simpson AJ, Old LJ. The cancer/testis genes: review, standardization, and commentary. Cancer Immun. 2004;4:1. [PubMed] [Google Scholar]
  • 27.Postovit LM, et al. The commonality of plasticity underlying multipotent tumor cells and embryonic stem cells. J Cell Biochem. 2007;101:908–917. doi: 10.1002/jcb.21227. [DOI] [PubMed] [Google Scholar]
  • 28.Ben-Porath I, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008;40:499–507. doi: 10.1038/ng.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kim J, et al. A Myc network accounts for similarities between embryonic stem and cancer cell transcription programs. Cell. 2010;143:313–324. doi: 10.1016/j.cell.2010.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ma YL, et al. Human embryonic stem cells and metastatic colorectal cancer cells shared the common endogenous human microRNA-26b. J Cell Mol Med. 2011;15:1941–1954. doi: 10.1111/j.1582-4934.2010.01170.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Stevenson BJ, et al. Rapid evolution of cancer/testis genes on the X chromosome. BMC Genomics. 2007;8:129. doi: 10.1186/1471-2164-8-129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Caballero OL, Chen YT. Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci. 2009;100:2014–2021. doi: 10.1111/j.1349-7006.2009.01303.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Scanlan MJ, Gure AO, Jungbluth AA, Old LJ, Chen YT. Cancer/testis antigens: an expanding family of targets for cancer immunotherapy. Immunol Rev. 2002;188:22–32. doi: 10.1034/j.1600-065x.2002.18803.x. [DOI] [PubMed] [Google Scholar]
  • 34.Almeida LG, et al. CTdatabase: a knowledgebase of high-throughput and curated data on cancer-testis antigens. Nucleic Acids Res. 2009;37:816–819. doi: 10.1093/nar/gkn673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Johns Hopkins University School of Medicine. ACTAbase: a comprehensive database for cancer/testis antigens. 2011 [online], http://actabase.jhu.edu.
  • 36.Andrade VC, et al. Prognostic impact of cancer/testis antigen expression in advanced stage multiple myeloma patients. Cancer Immun. 2008;8:2. [PMC free article] [PubMed] [Google Scholar]
  • 37.Schroder FH, et al. Screening and prostatecancer mortality in a randomized European study. N Engl J Med. 2009;360:1320–1328. doi: 10.1056/NEJMoa0810084. [DOI] [PubMed] [Google Scholar]
  • 38.Andriole GL, et al. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med. 2009;360:1310–1319. doi: 10.1056/NEJMoa0810696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Draisma G, et al. Lead time and overdiagnosis in prostate-specific antigen screening: importance of methods and context. J Natl Cancer Inst. 2009;101:374–383. doi: 10.1093/jnci/djp001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Fossa A, et al. NY-ESO-1 protein expression and humoral immune responses in prostate cancer. Prostate. 2004;59:440–447. doi: 10.1002/pros.20025. [DOI] [PubMed] [Google Scholar]
  • 41.Hudolin T, et al. Immunohistochemical expression of tumor antigens MAGE-A1, MAGE-A3/4, and NY-ESO-1 in cancerous and benign prostatic tissue. Prostate. 2006;66:13–18. doi: 10.1002/pros.20312. [DOI] [PubMed] [Google Scholar]
  • 42.Smith HA, Cronk RJ, Lang JM, McNeel DG. Expression and immunotherapeutic targeting of the SSX family of cancer-testis antigens in prostate cancer. Cancer Res. 2011;71:6785–6795. doi: 10.1158/0008-5472.CAN-11-2127. [DOI] [PubMed] [Google Scholar]
  • 43.Shiraishi T, et al. Cancer/testis antigens as potential predictors of biochemical recurrence of prostate cancer following radical prostatectomy. J Transl Med. 2011;9:153. doi: 10.1186/1479-5876-9-153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Xie C, et al. A novel multiplex assay combining autoantibodies plus PSA has potential implications for classification of prostate cancer from non-malignant cases. J Transl Med. 2011;9:43. doi: 10.1186/1479-5876-9-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Szmania S, Tricot G, van Rhee F. NY-ESO-1 immunotherapy for multiple myeloma. Leuk Lymphoma. 2006;47:2037–2048. doi: 10.1080/10428190600742292. [DOI] [PubMed] [Google Scholar]
  • 46.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]
  • 47.Nicholaou T, et al. Regulatory T-cell-mediated attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vaccine in patients with advanced malignant melanoma. Clin Cancer Res. 2009;15:2166–2173. doi: 10.1158/1078-0432.CCR-08-2484. [DOI] [PubMed] [Google Scholar]
  • 48.US National Library of Medicine. ClinicalTrials. gov. 2011 [online], http://clinicaltrials.gov/ct2/show/NCT00711334.
  • 49.Dubovsky JA, McNeel DG. Inducible expression of a prostate cancer-testis antigen, SSX-2, following treatment with a DNA methylation inhibitor. Prostate. 2007;67:1781–1790. doi: 10.1002/pros.20665. [DOI] [PubMed] [Google Scholar]
  • 50.Smith HA, McNeel DG. Vaccines targeting the cancer-testis antigen SSX-2 elicit HLA-A2 epitope-specific cytolytic T cells. J Immunother. 2011;34:569–580. doi: 10.1097/CJI.0b013e31822b5b1d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yokokawa J, et al. Identification of cytotoxic T-lymphocyte epitope(s) and its agonist epitope(s) of a novel target for vaccine therapy (PAGE4) Int J Cancer. 2007;121:595–605. doi: 10.1002/ijc.22698. [DOI] [PubMed] [Google Scholar]
  • 52.Parmigiani RB, et al. Characterization of a cancer/testis (CT) antigen gene family capable of eliciting humoral response in cancer patients. Proc Natl Acad Sci USA. 2006;103:18066–18071. doi: 10.1073/pnas.0608853103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Parmigiani RB, et al. Antibodies against the cancer-testis antigen CTSP-1 are frequently found in prostate cancer patients and are an independent prognostic factor for biochemical-recurrence. Int J Cancer. 2008;122:2385–2390. doi: 10.1002/ijc.23369. [DOI] [PubMed] [Google Scholar]
  • 54.Miles AK, et al. Identification of a novel prostate cancer-associated tumor antigen. Prostate. 2007;67:274–287. doi: 10.1002/pros.20520. [DOI] [PubMed] [Google Scholar]
  • 55.Chiriva-Internati M, et al. Identification of AKAP-4 as a new cancer/testis antigen for detection and immunotherapy of prostate cancer. Prostate. 2012;72:12–23. doi: 10.1002/pros.21400. [DOI] [PubMed] [Google Scholar]
  • 56.Fong L, et al. Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res. 2009;69:609–615. doi: 10.1158/0008-5472.CAN-08-3529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Karpf AR, Bai S, James SR, Mohler JL, Wilson EM. Increased expression of androgen receptor coregulator MAGE-11 in prostate cancer by DNA hypomethylation and cyclic AMP. Mol Cancer Res. 2009;7:523–535. doi: 10.1158/1541-7786.MCR-08-0400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bai S, He B, Wilson EM. Melanoma antigen gene protein MAGE-11 regulates androgen receptor function by modulating the interdomain interaction. Mol Cell Biol. 2005;25:1238–1257. doi: 10.1128/MCB.25.4.1238-1257.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wilson EM. Androgen receptor molecular biology and potential targets in prostate cancer. Ther Adv Urol. 2010;2:105–117. doi: 10.1177/1756287210372380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Cronwright G, et al. Cancer/testis antigen expression in human mesenchymal stem cells: down-regulation of SSX impairs cell migration and matrix metalloproteinase 2 expression. Cancer Res. 2005;65:2207–2215. doi: 10.1158/0008-5472.CAN-04-1882. [DOI] [PubMed] [Google Scholar]
  • 61.Xiang Y, et al. JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer. Proc Natl Acad Sci USA. 2007;104:19226–19231. doi: 10.1073/pnas.0700735104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kaufman DS, Shipley WU, Feldman AS. Bladder cancer. Lancet. 2009;374:239–249. doi: 10.1016/S0140-6736(09)60491-8. [DOI] [PubMed] [Google Scholar]
  • 63.Tanaka MF, Sonpavde G. Diagnosis and management of urothelial carcinoma of the bladder. Postgrad Med. 2011;123:43–55. doi: 10.3810/pgm.2011.05.2283. [DOI] [PubMed] [Google Scholar]
  • 64.Babjuk M, et al. EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder. Eur Urol. 2008;54:303–314. doi: 10.1016/j.eururo.2008.04.051. [DOI] [PubMed] [Google Scholar]
  • 65.Herr HW, et al. Neoadjuvant chemotherapy in invasive bladder cancer: the evolving role of surgery. J Urol. 1990;144:1083–1088. doi: 10.1016/s0022-5347(17)39664-7. [DOI] [PubMed] [Google Scholar]
  • 66.Patard JJ, et al. Expression of MAGE genes in transitional-cell carcinomas of the urinary bladder. Int J Cancer. 1995;64:60–64. doi: 10.1002/ijc.2910640112. [DOI] [PubMed] [Google Scholar]
  • 67.Jungbluth AA, et al. Expression of MAGE-antigens in normal tissues and cancer. Int J Cancer. 2000;85:460–465. [PubMed] [Google Scholar]
  • 68.Bergeron A, et al. High frequency of MAGE-A4 and MAGE-A9 expression in high-risk bladder cancer. Int J Cancer. 2009;125:1365–1371. doi: 10.1002/ijc.24503. [DOI] [PubMed] [Google Scholar]
  • 69.Schultz-Thater E, et al. 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]
  • 70.Chen YT, et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci USA. 1997;94:1914–1918. doi: 10.1073/pnas.94.5.1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bolli M, et al. NY-ESO-1/LAGE-1 coexpression with MAGE-A cancer/testis antigens: a tissue microarray study. Int J Cancer. 2005;115:960–966. doi: 10.1002/ijc.20953. [DOI] [PubMed] [Google Scholar]
  • 72.Ono T, et al. Identification of proacrosin binding protein sp32 precursor as a human cancer/testis antigen. Proc Natl Acad Sci USA. 2001;98:3282–3287. doi: 10.1073/pnas.041625098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Matsuda R, et al. LY6K is a novel molecular target in bladder cancer on basis of integrate genome-wide profiling. Br J Cancer. 2011;104:376–386. doi: 10.1038/sj.bjc.6605990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hayami S, et al. Overexpression of the JmjC histone demethylase KDM5B in human carcinogenesis: involvement in the proliferation of cancer cells through the E2F/RB pathway. Mol Cancer. 2010;9:59. doi: 10.1186/1476-4598-9-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Nishiyama T, et al. Immunotherapy of bladder cancer using autologous dendritic cells pulsed with human lymphocyte antigen-A24-specific MAGE-3 peptide. Clin Cancer Res. 2001;7:23–31. [PubMed] [Google Scholar]
  • 76.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]
  • 77.Parkin CM, Whelan SL, Ferlay J, Teppo L, Thomas D. IARC Scientific Publications No. 155. International Agency for Research on Cancer; Lyon, France: 2002. [Google Scholar]
  • 78.National Cancer Institute. SEER Program Public Use Data Tapes 1973–2002. 2005. Surveillance Research Program; Cancer Statistics Branch. [Google Scholar]
  • 79.Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. N Engl J Med. 1996;335:865–875. doi: 10.1056/NEJM199609193351207. [DOI] [PubMed] [Google Scholar]
  • 80.Karakiewicz PI, et al. Multi-institutional validation of a new renal cancer-specific survival nomogram. J Clin Oncol. 2007;25:1316–1322. doi: 10.1200/JCO.2006.06.1218. [DOI] [PubMed] [Google Scholar]
  • 81.Lam JS, Leppert JT, Figlin RA, Belldegrun AS. Role of molecular markers in the diagnosis and therapy of renal cell carcinoma. Urology. 2005;66:1–9. doi: 10.1016/j.urology.2005.06.112. [DOI] [PubMed] [Google Scholar]
  • 82.Zini L, et al. Population-based assessment of survival after cytoreductive nephrectomy versus no surgery in patients with metastatic renal cell carcinoma. Urology. 2009;73:342–346. doi: 10.1016/j.urology.2008.09.022. [DOI] [PubMed] [Google Scholar]
  • 83.Yagoda A. Phase II cytotoxic chemotherapy trials in renal cell carcinoma: 1983–1988. Prog Clin Biol Res. 1990;350:227–241. [PubMed] [Google Scholar]
  • 84.Motzer RJ, Russo P. Systemic therapy for renal cell carcinoma. J Urol. 2000;163:408–417. [PubMed] [Google Scholar]
  • 85.Jungbluth AA, et al. 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]
  • 86.Yamanaka K, et al. Expression of MAGE genes in renal cell carcinoma. Int J Mol Med. 1998;2:57–60. doi: 10.3892/ijmm.2.1.57. [DOI] [PubMed] [Google Scholar]
  • 87.Garg M, et al. Sperm-associated antigen 9 is associated with tumor growth, migration, and invasion in renal cell carcinoma. Cancer Res. 2008;68:8240–8248. doi: 10.1158/0008-5472.CAN-08-1708. [DOI] [PubMed] [Google Scholar]
  • 88.Bukowski RM. Immunotherapy in renal cell carcinoma. Oncology (Williston Park) 1999;13:801–813. [PubMed] [Google Scholar]
  • 89.Schmidinger M, et al. Sequential administration of interferon gamma and interleukin-2 in metastatic renal cell carcinoma: results of a phase II trial. Austrian Renal Cell Carcinoma Study Group. Cancer Immunol Immunother. 2000;49:395–400. doi: 10.1007/s002620000128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Coral S, et al. 5-aza-2′-deoxycytidine-induced expression of functional cancer testis antigens in human renal cell carcinoma: immunotherapeutic implications. Clin Cancer Res. 2002;8:2690–2695. [PubMed] [Google Scholar]
  • 91.Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012;62:10–29. doi: 10.3322/caac.20138. [DOI] [PubMed] [Google Scholar]
  • 92.Rosen A, Jayram G, Drazer M, Eggener SE. Global trends in testicular cancer incidence and mortality. Eur Urol. 2011;60:374–379. doi: 10.1016/j.eururo.2011.05.004. [DOI] [PubMed] [Google Scholar]
  • 93.Feldman DR, Bosl GJ, Sheinfeld J, Motzer RJ. Medical treatment of advanced testicular cancer. JAMA. 2008;299:672–684. doi: 10.1001/jama.299.6.672. [DOI] [PubMed] [Google Scholar]
  • 94.Oosterhuis JW, Looijenga LH. Testicular germ-cell tumours in a broader perspective. Nat Rev Cancer. 2005;5:210–222. doi: 10.1038/nrc1568. [DOI] [PubMed] [Google Scholar]
  • 95.Jacobsen GK. Alpha-fetoprotein (AFP) and human chorionic gonadotropin (HCG) in testicular germ cell tumours. A comparison of histologic and serologic occurrence of tumour markers. Acta Pathol Microbiol Immunol Scand A. 1983;91:183–190. doi: 10.1111/j.1699-0463.1983.tb02744.x. [DOI] [PubMed] [Google Scholar]
  • 96.Rajpert-De Meyts E. Developmental model for the pathogenesis of testicular carcinoma in situ: genetic and environmental aspects. Hum Reprod Update. 2006;12:303–323. doi: 10.1093/humupd/dmk006. [DOI] [PubMed] [Google Scholar]
  • 97.Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448:313–317. doi: 10.1038/nature05934. [DOI] [PubMed] [Google Scholar]
  • 98.Geldmacher A, Freier A, Losch FO, Walden P. Therapeutic vaccination for cancer immunotherapy: antigen selection and clinical responses. Hum Vaccin. 2011;7:S115–S119. doi: 10.4161/hv.7.0.14573. [DOI] [PubMed] [Google Scholar]
  • 99.Fratta E, et al. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol Oncol. 2011;5:164–182. doi: 10.1016/j.molonc.2011.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Garcia-Manero G. Treatment of higher-risk myelodysplastic syndrome. Semin Oncol. 2011;38:673–681. doi: 10.1053/j.seminoncol.2011.07.001. [DOI] [PubMed] [Google Scholar]
  • 101.Coral S, et al. Prolonged upregulation of the expression of HLA class I antigens and costimulatory molecules on melanoma cells treated with 5-aza-2′-deoxycytidine (5-AZA-CdR) J Immunother. 1999;22:16–24. doi: 10.1097/00002371-199901000-00003. [DOI] [PubMed] [Google Scholar]
  • 102.Penney KL, et al. mRNA expression signature of Gleason grade predicts lethal prostate cancer. J Clin Oncol. 2011;29:2391–2396. doi: 10.1200/JCO.2010.32.6421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Cuzick J, et al. Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study. Lancet Oncol. 2011;12:245–255. doi: 10.1016/S1470-2045(10)70295-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kulkarni MM. Chapter 25: digital multiplexed gene expression analysis using the NanoString nCounter system. Greene Pub. Associates; Brooklyn, NY: 2011. [DOI] [PubMed] [Google Scholar]
  • 105.Cheung IY, Cheung NK. Molecular detection of GAGE expression in peripheral blood and bone marrow: utility as a tumor marker for neuroblastoma. Clin Cancer Res. 1997;3:821–826. [PubMed] [Google Scholar]
  • 106.Lu Y, Wu LQ, Lu ZH, Wang XJ, Yang JY. Expression of SSX-1 and NY-ESO-1 mRNA in tumor tissues and its corresponding peripheral blood expression in patients with hepatocellular carcinoma. Chin Med J (Engl) 2007;120:1042–1046. [PubMed] [Google Scholar]
  • 107.de Bono JS, et al. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2008;14:6302–6309. doi: 10.1158/1078-0432.CCR-08-0872. [DOI] [PubMed] [Google Scholar]
  • 108.Rajagopalan K, Mooney SM, Parekh N, Getzenberg RH, Kulkarni P. A majority of the cancer/testis antigens are intrinsically disordered proteins. J Cell Biochem. 2011;112:3256–3267. doi: 10.1002/jcb.23252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Vavouri T, Semple JI, Garcia-Verdugo R, Lehner B. Intrinsic protein disorder and interaction promiscuity are widely associated with dosage sensitivity. Cell. 2009;138:198–208. doi: 10.1016/j.cell.2009.04.029. [DOI] [PubMed] [Google Scholar]
  • 110.Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–210. doi: 10.1093/nar/30.1.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.University of Auckland. R: a language and environment for statistical computing. 2010 [online], http://www.R-project.org.
  • 112.Gentleman RC, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5:80. doi: 10.1186/gb-2004-5-10-r80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.University of Rochester Medical Center. The Comprehensive R Archive Network. [online], http://cran.r-project.org/web/packages/gplots/index.html.

RESOURCES