Next Article in Journal
Recent Advances in Imaging Macular Atrophy for Late-Stage Age-Related Macular Degeneration
Previous Article in Journal
Hereditary Transthyretin Amyloidosis: How to Differentiate Carriers and Patients Using Speckle-Tracking Echocardiography
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Expression of Preferentially Expressed Antigen in Melanoma, a Cancer/Testis Antigen, in Carcinoma In Situ of the Urinary Tract

1
Department of Pathology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
2
Department of Urology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
3
Translational Research Program, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
4
Department of Thoracic Surgery, Kansai Medical University, 2-5-1, Shinmachi, Hirakata 573-1010, Osaka, Japan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2023, 13(24), 3636; https://doi.org/10.3390/diagnostics13243636
Submission received: 8 November 2023 / Revised: 7 December 2023 / Accepted: 9 December 2023 / Published: 10 December 2023

Abstract

:
Carcinoma in situ (CIS) of the urinary tract comprises 1–3% of all urothelial malignancies and is often a precursor to muscle-invasive urothelial carcinoma (UC). This study aimed to examine the expression profiles of preferentially expressed antigen in melanoma (PRAME), a cancer/testis antigen, and assess its diagnostic and therapeutic applications in CIS, given that its expression in UC has been minimally studied and has not yet been analyzed in CIS. We selected consecutive patients with CIS who underwent biopsy and/or transurethral tumor resection at the Osaka Medical and Pharmaceutical University Hospital. Immunohistochemical staining for PRAME and p53 was performed. Overall, 53 patients with CIS (6 females and 47 males) were included. Notably, PRAME expression was observed in 23 of the 53 patients (43.4%), whereas it was absent in the non-neoplastic urothelial epithelium. Furthermore, no correlation was found between PRAME expression and aberrant p53 expression. Therefore, PRAME expression may serve as a useful marker for CIS of the urinary tract. Furthermore, PRAME may be a candidate for the novel therapeutic target for standard treatment-refractory CIS patients.

1. Introduction

Urothelial carcinoma (UC) is one of the common causes of cancer-related death worldwide. Most UCs show papillary proliferation, while carcinoma in situ (CIS) is defined as a flat neoplastic proliferation of high-grade neoplastic urothelial cells without papillary formation, comprising approximately 1–3% of all urothelial malignancies [1]. Notably, CIS is considered to carry a high risk for the development of muscle-invasive UC, as over 80% of untreated patients with CIS progress to muscle-invasive UC [2]. Therefore, early detection and treatment of CIS are among the important issues in the field of urological oncology.
As CIS does not form papillary tumors, the standard method for diagnosing CIS of the urinary tract is biopsy from all suspicious erythematous areas and/or random biopsies from the normal-appearing mucosa by cystoscopic examinations [2]. Furthermore, urine cytology has been recognized as the standard and valuable diagnostic tool for detecting high-grade UC and CIS [2] because it represents the highest predictive value in diagnosing high-grade UC. Importantly, one report demonstrated that the urine cytological examination showed a sensitivity and specificity of 87.1% and 63%, respectively, in patients with CIS [3]. Another study showed a sensitivity of 54.5% using urine cytology for detecting CIS [4]. Therefore, a significant number of patients with CIS yield negative results in cytological examinations. Accordingly, several ancillary methods, including fluorescence in situ hybridization, have been developed either alone or in combination with urine cytological examination to improve the detection of carcinoma cells. Moreover, recent studies demonstrated the usefulness of a DNA methylation test for detecting high-grade UC [5,6,7]. Reportedly, the bladder EpiCheck test® analyzed the methylation status of 15 selected genes using urine specimens, and showed a high sensitivity for detecting high-grade UC [5,6,7]. Although some of these markers aid in improving the sensitivity of detection of carcinoma cells, they have not yet been universally used because of the cost and false-negative results [2,5]. Therefore, the development of a novel and convenient diagnostic marker is required to detect UC cells, especially CIS.
Preferentially expressed antigen in melanoma (PRAME) is one of the cancer/testis antigens. These antigens are a family of tumor-associated antigens expressed in malignant tumors but not in normal adult tissues, except for the testis. PRAME was first reported as a tumor-associated antigen expressed in malignant melanoma recognized by cytotoxic T lymphocytes [8]. The PRAME gene is located on chromosome 22 (22q11.22) and contains leucine-rich repeat domains [9]. This protein acts as a repressor of the retinoic acid receptor pathway, leading to the regulation of cell growth, differentiation, and apoptosis [10,11,12]. Importantly, its expression is fundamentally restricted in the non-neoplastic testis, specifically spermatogonia, and proliferative endometrial glands; it is regulated at very low levels by DNA methylation in most of the non-neoplastic tissues excluding the testis and endometrium [8,13]. Recently, PRAME expression has received much attention in some types of malignant tumor as a useful diagnostic marker due to its nature of being a cancer/testis antigen [13]. Notably, malignant melanoma and its mimics represents the most intensively investigated area of PRAME expression [14,15,16,17,18,19,20,21,22,23,24]. Numerous articles have examined the expression profiles of PRAME in malignant melanoma and have demonstrated its diagnostic utility. Specifically, most cases of malignant melanoma exhibited positive immunoreactivity to PRAME; in contrast, most benign melanocytic nevi did not [14,15,16,17,18,19,20,21,22,23,24]. Moreover, PRAME expression has also been reported in various types of malignant tumor, other than malignant melanoma, including myxoid liposarcoma, synovial sarcoma, neuroblastoma, renal cell carcinoma, non-small-cell lung cancer, and thymic cancer [8,13,14,25,26,27,28,29,30]. The expression of PRAME in UC has been analyzed in only three studies, wherein it was reported in 7%, 15%, and 20% of patients with UC, as determined by immunohistochemistry or RT-PCR [13,31,32]. However, the expression profiles of PRAME in CIS of the urinary tract have not yet been examined.
Furthermore, due to the nature of the cancer/testis antigens of PRAME, this protein has been recognized as a potential target for immunotherapy in patients with some types of malignant tumor, such as uveal malignant melanoma and non-small-cell lung cancer [33,34]. Gezgin et al. showed that PRAME-specific T cells reacted against PRAME-positive uveal malignant melanoma cell lines, and approximately 45% of primary uveal malignant melanoma showed positive immunoreactivity for PRAME [33]. Thus, PRAME may be a potential candidate for direct immunotherapy for patients with uveal malignant melanoma expressing PRAME [33]. In another study, a phase I dose escalation study for patients with PRAME-positive resected non-small-cell lung cancer was performed [34]. Recombinant PRAME proteins were injected for measuring the anti-PRAME humoral response in the study, and PRAME-specific CD4-positive T cells were detected after immunizations in all patients, but not CD8-positive T cells [34]. Only low-grade adverse events, such as injection site reactions and fever, were observed; therefore, PRAME might be a promising candidate for immunotherapy for patients with non-small-cell lung cancer [34]. These results indicate that PRAME may be a potential immunotherapeutic target for all patients with PRAME-expressing malignant tumors, independent of histological types. However, the PRAME expression profiles of UC have not yet been analyzed in detail, and it has not been elucidated whether or not PRAME is also a potential target for immunotherapy for patients with UC, especially CIS.
Accordingly, the present study aimed to analyze the immunohistochemical expression profiles of PRAME in CIS of the urinary tract and discuss the potential diagnostic and therapeutic applications.

2. Materials and Methods

2.1. Patient Selection

We selected consecutive patients with CIS who underwent biopsy and/or transurethral/surgical resection at the Osaka Medical and Pharmaceutical University Hospital between January 2021 and December 2022. Accordingly, 53 patients were included in this study.
This retrospective, single-institution study was conducted according to the principles of the Declaration of Helsinki, and the study protocol was approved by the Institutional Review Board of Osaka Medical and Pharmaceutical University (Approval #2020-124 and #2023-027). All data were anonymized. The institutional review board waived the requirement for informed consent because of the retrospective study design, as medical records and archived samples were used with no risk to the participants. Moreover, the present study did not include minors. Information regarding this study, such as the inclusion criteria and the opportunity to opt out, was provided through the institutional website (https://www.ompu.ac.jp/u-deps/path/img/file9.pdf) (accessed on 7 December 2023).

2.2. Histopathological Analysis

Biopsied and/or resected specimens were fixed with 10% buffered formalin, sectioned, and stained with hematoxylin and eosin. A review of the histopathological features was performed by two researchers (SF and MI). The diagnostic criteria for CIS were based on the descriptions provided in the WHO Classification of Tumours [1]. Cases showing the typical histopathological features of CIS were selected, and questionable cases of patients with reactive atypia or dysplasia were excluded from this study. Moreover, cases of invasive UC featuring a CIS component were excluded from this study.

2.3. Immunohistochemical Analyses

Immunohistochemical staining was performed using autostainers (Discovery Ultra System; Roche Diagnostics, Basel, Switzerland; Leica Bond-III; Leica Biosystems GmbH, Nußloch, Germany) according to the manufacturer’s instructions. A mouse monoclonal antibody against p53 (DO-7; DAKO; Agilent Technologies, Inc., Santa Clara, CA, USA, diluted 1:50) and a rabbit monoclonal antibody against PRAME (EPR20330; Abcam, Cambridge, UK, diluted 1:100) were used. Whole biopsied or resected specimens of CIS were used for immunostaining, and no tissue microarray was applied in the present study. A minimum of two researchers independently evaluated the immunohistochemical staining results.
Positive immunoreactivity for PRAME, manifesting as either nuclear and/or cytoplasmic positivity, was defined as any expression in carcinoma cells of CIS, employing the same methodology as that outlined in the previous report [31]. Similarly, p53 immunoreactivity was assessed in accordance with the previous report [35], as follows: aberrant patterns included either diffuse positive (with the majority of carcinoma cells exhibiting homogeneous moderate to strong positivity) or negative/null patterns, while wild-type patterns featured patchy positive cells.

2.4. Statistical Analysis

The correlation between the two groups was analyzed using Fisher’s exact test. A p-value < 0.05 was considered statistically significant.

3. Results

3.1. Patient Characteristics

This study included 53 patients (6 female and 47 male). The median age was 73 years (range: 47–92 years). The lesion locations were the urinary bladder and ureter in 50 and 3 patients, respectively.

3.2. Histopathological and Immunohistochemical Features

Typical histopathological features of CIS are shown in Figure 1. Flat proliferation of the neoplastic urothelial cells with large round to oval nuclei with or without conspicuous nucleoli was noted. The nuclei size was more than three or four times larger than that of the non-neoplastic urothelial cells, and these nuclei showed an irregular contour and hyperchromasia. Mitotic figures were observed in most cases. No invasive neoplastic growth was observed in any of the lesions.
Figure 2 shows a typical immunohistochemical expression for PRAME in CIS. PRAME expression was noted in 23 of 53 patients (43.4%), but the non-neoplastic urothelial epithelia around the CIS lesions showed no positive immunoreactivity for PRAME in all cases (p < 0.001). Positive immunoreactivity for PRAME in CIS was noted in the nuclei and/or cytoplasm of the neoplastic cells of CIS, mostly only in the nuclei of the neoplastic cells. Aberrant and wild-type patterns of p53 expression are shown in Figure 3.
Aberrant p53 expression (diffuse positive or null patterns) was noted in 29 patients (54.7%) and a wild-type pattern was noted in 24 patients (45.3%).
No correlation was noted between PRAME expression and p53 expression patterns (Table 1, p = 0.82).

4. Discussion

The expression of PRAME has been reported in various types of malignant tumor [8,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30]. Notably, the present study is the first study to clearly demonstrate that PRAME was expressed in 43.4% of CIS of the urinary tract. Only three previous studies addressed the expression profiles of PRAME in UC. Table 2 summarizes the expression profiles of PRAME expression in UC of the previous studies, as well as that of the present study. An immunohistochemical study using tissue microarrays revealed that only 7% of UC (the information regarding histological grade and whether it was invasive or non-invasive were not available) showed positive immunoreactivity for PRAME [13]. In another immunohistochemical study using tissue microarrays, PRAME expression was noted in 15% of muscle-invasive high-grade UC [31]. Furthermore, an RT-PCR assay showed PRAME expression in 20% of non-invasive papillary and invasive UC cases [32]. Importantly, PRAME expression was observed in only 3% of non-invasive papillary UC, compared to 16% in pT1 invasive UC and 28% in pT2-4 invasive UC [32]. Moreover, although a significant correlation was found between the presence of CIS and PRAME expression in patients with non-muscle invasive UC, information regarding PRAME expression, specifically regarding the CIS component, was not available [32]. According to the results of these previous studies, PRAME expression is not a rare finding, especially in muscle-invasive and higher histological grade UC, although the detection methods and primary antibody used in immunohistochemical analyses were not the same [13,31,32]. In the present study, we demonstrated that 43.4% of CIS of the urinary tract showed positive immunoreactivity for PRAME, which was a rate higher than that reported for muscle-invasive UC and non-invasive papillary UC [31,32]. This result suggests that the expression of PRAME may differ between papillary UC and CIS, and might be related to carcinogenesis of CIS of the urinary tract. Moreover, this difference might be related to the relationship between PRAME expression and presence of CIS in patients with non-muscle invasive UC [32].
PRAME expression is closely regulated at very low levels by DNA methylation in adult non-neoplastic tissues except the testis and endometrium [8,13]. Due to its nature as a cancer/testis antigen, PRAME expression has been recognized as a useful diagnostic marker for several types of malignant tumor, especially malignant melanomas [14,15,16,17,18,19,20,21,22,23,24]. Table 3 summarizes the typical examples of PRAME expression in the various types of non-epithelial and epithelial malignant tumors. A high frequency of PRAME expression is reported in malignant melanomas, myxoid liposarcomas, synovial sarcomas, neuroblastomas, and thymic squamous cell carcinomas [13,25,26,27,28,30,36]. PRAME expression is more frequently noted in non-epithelial malignant tumors than in epithelial malignant tumors (Table 3); moreover, its expression has been noted in various types of non-epithelial and epithelial malignant tumors at low-to-intermediate levels (Table 3) [13,25,36,37,38,39,40,41,42,43,44,45,46,47,48]. Importantly, no PRAME expression was reported in some types of malignant tumor, such as prostate cancer, gastric cancer, and gastrointestinal stromal tumors [13]. Therefore, PRAME expression might depend on the origin of the malignant tumors. PRAME expression can be readily detected using the immunohistochemical method, as demonstrated in this study, and it may be particularly useful for differentiating malignant tumors, notably malignant melanomas, from benign melanocytic nevi. However, the positive ratio of PRAME varies according to the tumor’s origin. Therefore, the origin of the tumor must be considered when PRAME expression is used in the diagnosis of malignant tumors.
The mechanism of PRAME expression in malignant tumors has not yet been elucidated in detail. PRAME expression is reported to be regulated by some genes, such as SOX17 (SRY-box 17) and MZF1 (myeloid zing finger 1) [12]; moreover, it shows its functions via regulation of some target genes, including p53, p21, retinoic acid receptor, and S100A4, leading to cell proliferation control, differentiation, growth arrest, and apoptosis in several human malignancies [12]. For example, PRAME expression is promoted by MZF-1, which leads to the enhancement of colony-forming capability of melanoma cells, and is inhibited by miR-211 in malignant melanoma cells [12,49]. PRAME is downstream of SOX17 and LIN28 for regulating pluripotency and controlling germ cell differentiation in seminoma cells [50]. In acute myeloid leukemia cells, the overexpression of PRAME correlates with the decreased expression of Hsp27 (heat shock protein 27), S100A4, and p21 at the transcription levels [51]. Moreover, PRAME expression is associated with the cell cycle from the G0/G1 phase to the S phase, and its overexpression correlates with the suppression of apoptosis in leukemic cells [52]. Accordingly, PRAME expression may control cell proliferation, differentiation, and apoptosis via the several above-mentioned molecular pathways in some types of malignant tumor. However, detailed molecular mechanisms regarding controlling PRAME expression and its downstream pathways remains unclear; moreover, these mechanisms might be different depending the origin of malignant tumors. Recently, it has been reported that PRAME forms a complex with p14/alternate reading frame (ARF) (CDKN2A), a well-established tumor suppressor, as well as with Cullin 2 RING E3 ligases [53]. Specifically, PRAME serves as a specific receptor protein for targeting p14/ARF for degradation, and this complex consequently inhibits cell growth [53]. Notably, the downregulation of PRAME significantly impedes cancer cell growth by inducing G2/M cell cycle arrest in hepatocellular carcinoma cell lines and lung cancer cell lines [50]. This molecular mechanism may elucidate the relationship between PRAME expression and the malignant nature of tumors. However, the detailed molecular mechanisms governing the upstream signaling pathways of PRAME expression in UC, especially CIS, remain to be elucidated. Further studies are required to clarify this mechanism.
Further, the present study demonstrated that PRAME expression in CIS of the urinary tract did not correlate with p53 aberrant expression. Diffuse positive or null immunoreactivity for p53 has been considered to be one of the characteristic features of CIS of the urinary tract and, thus, has been used as a useful diagnostic marker in the setting of pathological diagnosis [35,54]. However, recent studies have demonstrated that a relatively large number of CIS cases showed wild-type expression patterns of p53 (approximately 30–60% of CIS in the previous reports and 45.3% of the present cohort) [35,54]. In the present cohort, 10 of 24 (41.7%) p53 wild-type CIS patients showed positive immunoreactivity for PRAME, and expression patterns of p53 were not used for diagnosis of CIS in these patients. Therefore, immunohistochemical staining for PRAME, especially a combination of PRAME and p53, might help to diagnose CIS of the urinary tract.
The current therapeutic options for patients with CIS are limited, and intravesical Bacillus Calmette–Guérin (BCG) treatment has been the mainstay for this subset of patients [2]. However, a modest rate of complete response to BCG has been identified as the unmet need for innovative therapeutic approaches, as such patients would be candidates for radical cystectomy [2]. Although the efficacy of novel immunotherapeutic or viral agents, including anti-PD-1 inhibitors and nadofaragene firadenovec, has been investigated for these patients [55,56,57], developing new therapeutic targets has been an urgent issue. Recently, PRAME has garnered considerable attention for its therapeutic potential in the field of oncology [11]. This protein is recognized as one of the potential targets for immunotherapy because it is a cancer/testis antigen, and the peptides on MHC class I derived from PRAME are recognized by cytotoxic T cells [8,12,33,34,58]. Additionally, some clinical studies have been performed to evaluate the usefulness of PRAME as a target for antigen-specific immunization by adoptive T-cell immunotherapy in patients with metastatic uveal malignant melanoma and non-small-cell lung cancer [33,34,58]. Moreover, PRAME expression has been reported to be correlated with chemotherapy resistance in patients with UC and some malignant lymphomas [12,32,38,59]. Thus, PRAME may serve as a candidate for immunotherapy in patients with high-grade UC, as well as in those with CIS.
There are several limitations to the present study. First, this was a single-center retrospective analysis with a relatively small number of CIS patients, potentially leading to bias in the statistical power. Therefore, additional multi-center studies with larger cohorts are needed. Second, the expression of PRAME was analyzed by the immunohistochemical method, employing the same method and antibody as carried out in a previous report [31]. RT-PCR was used for the detection of PRAME expression in UC in another study [32]. Moreover, the correlation between protein expression detected by the immunohistochemical method and mRNA expression by RT-PCR was not available in the present study. However, a significant positive correlation between immunohistochemical analysis and PCR measurement of PRAME expression in malignant lymphoma has been shown [38]. Thus, the immunohistochemical analysis may be useful and convenient for detecting PRAME expression.

5. Conclusions

The present study demonstrated that 43.4% of CIS of the urinary tract showed positive immunoreactivity for PRAME. Detection of PRAME may be a useful marker for CIS of the urinary tract due to the nature of the cancer/testis antigen and might be a candidate for immunotherapy for CIS patients. Further studies are needed to clarify the expression profiles of this protein in the larger cohort. Moreover, the application of PRAME expression by the less invasive diagnostic methods for CIS compared to biopsy must be studied.

Author Contributions

Conception and design of the study: S.F. and M.I.; histopathological and immunohistochemical analyses: S.F. and M.I.; acquisition and analysis of data: S.F., M.I., K.K., K.N., T.T., T.S., Y.T., T.M., H.A. and Y.H.; drafting of the manuscript, tables, and figures: S.F. and M.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The present study was conducted in accordance with the Declaration of Helsinki, and the study protocol was approved by the Institutional Review Board of the Osaka Medical and Pharmaceutical University (protocol no. 2020-124 and 2023-027; Takatsuki, Osaka, Japan) (26 April 2023). All data are completely anonymized. Due to the retrospective nature of the present study, it was difficult to obtain informed consent from all subjects. Therefore, an opt-out approach was adopted based on the approval of the Ethics Committee of the Osaka Medical and Pharmaceutical University.

Informed Consent Statement

The institutional review board waived the requirement for informed consent because of the retrospective study design, as medical records and archived samples were used with no risk to the participants.

Data Availability Statement

All data generated and analyzed in this study are included in this published article.

Acknowledgments

The authors would like to express their gratitude to Shizuka Ono, Yusuke Ohnishi, and Naoto Kohno (Department of Pathology, Osaka Medical and Pharmaceutical University) for their technical assistance in this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Williamson, S.R.; Magi-Galluzzi, C.; Matoso, A.; Montironi, R.; Raspollini, M.R.; WHO Classification of Tumours. Urinary and Male Genital Tumours, 5th ed.; The WHO Classification of Tumors Editorial Board, Ed.; IARC: Lyon, France, 2022; pp. 147–149. [Google Scholar]
  2. Subiela, J.D.; Rodríguez Faba, O.; Guerrero Ramos, F.; Vila Reyes, H.; Pisano, F.; Breda, A.; Palou, J. Carcinoma in situ of the urinary bladder: A systematic review of current knowledge regarding detection, treatment, and outcomes. Eur. Urol. Focus 2020, 6, 674–682. [Google Scholar] [CrossRef]
  3. Subiela, J.D.; Palou, J.; Esquinas, C.; Fernández Gómez, J.M.; Rodríguez Faba, O. Clinical usefulness of random biopsies in diagnosis and treatment of non-muscle invasive bladder cancer: Systematic review and meta-analysis. Actas Urol. Esp. 2018, 42, 285–298. [Google Scholar] [CrossRef]
  4. Zang, Z.; Kesavan, N.R.; Esuvaranathan, K. UBC®Rapid Is Sensitive in Detecting High-Grade Bladder Urothelial Carcinoma and Carcinoma in situ in Asian Population. Urol. Int. 2023, 107, 29–34. [Google Scholar] [CrossRef] [PubMed]
  5. Pierconti, F.; Martini, M.; Fiorentino, V.; Cenci, T.; Capodimonti, S.; Straccia, P.; Sacco, E.; Pugliese, D.; Cindolo, L.; Larocca, L.M.; et al. The combination cytology/epichek test in non muscle invasive bladder carcinoma follow-up: Effective tool or useless expence? Urol. Oncol. 2021, 39, 131.e17–131.e21. [Google Scholar] [CrossRef] [PubMed]
  6. Pierconti, F.; Martini, M.; Fiorentino, V.; Cenci, T.; Racioppi, M.; Foschi, N.; Di Gianfrancesco, L.; Sacco, E.; Rossi, E.; Larocca, L.M.; et al. Upper urothelial tract high-grade carcinoma: Comparison of urine cytology and DNA methylation analysis in urinary samples. Hum. Pathol. 2021, 118, 42–48. [Google Scholar] [CrossRef] [PubMed]
  7. Peña, K.B.; Riu, F.; Hernandez, A.; Guilarte, C.; Badia, J.; Parada, D. Usefulness of the Urine Methylation Test (Bladder EpiCheck®) in Follow-Up Patients with Non-Muscle Invasive Bladder Cancer and Cytological Diagnosis of Atypical Urothelial Cells-An Institutional Study. J. Clin. Med. 2022, 11, 3855. [Google Scholar] [CrossRef]
  8. Ikeda, H.; Lethé, B.; Lehmann, F.; van Baren, N.; Baurain, J.F.; de Smet, C.; Chambost, H.; Vitale, M.; Moretta, A.; Boon, T.; et al. Characterization of an antigen that is recognized on a melanoma showing partial HLA loss by CTL expressing an NK inhibitory receptor. Immunity 1997, 6, 199–208. [Google Scholar] [CrossRef] [PubMed]
  9. Wadelin, F.; Fulton, J.; McEwan, P.A.; Spriggs, K.A.; Emsley, J.; Heery, D.M. Leucine-rich repeat protein PRAME: Expression, potential functions and clinical implications for leukaemia. Mol. Cancer 2010, 9, 226. [Google Scholar] [CrossRef] [PubMed]
  10. Epping, M.T.; Wang, L.; Edel, M.J.; Carlée, L.; Hernandez, M.; Bernards, R. The human tumor antigen PRAME is a dominant repressor of retinoic acid receptor signaling. Cell 2005, 122, 835–847. [Google Scholar] [CrossRef]
  11. Kern, C.H.; Yang, M.; Liu, W.S. The PRAME family of cancer testis antigens is essential for germline development and gametogenesis. Biol. Reprod. 2021, 105, 290–304. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, Y.; Zou, R.; Wang, J.; Wang, Z.W.; Zhu, X. The role of the cancer testis antigen PRAME in tumorigenesis and immunotherapy in human cancer. Cell Prolif. 2020, 53, e12770. [Google Scholar] [CrossRef]
  13. Kaczorowski, M.; Chłopek, M.; Kruczak, A.; Ryś, J.; Lasota, J.; Miettinen, M. PRAME expression in cancer. A systematic immuno-histochemical study of >5800 epithelial and nonepithelial tumors. Am. J. Surg. Pathol. 2022, 46, 1467–1476. [Google Scholar] [CrossRef]
  14. Lezcano, C.; Jungbluth, A.A.; Busam, K.J. Comparison of immunohistochemistry for PRAME with cytogenetic test results in the evaluation of challenging melanocytic tumors. Am. J. Surg. Pathol. 2020, 44, 893–900. [Google Scholar] [CrossRef]
  15. Kim, J.C.; Choi, J.W.; Kim, Y.C. Comparison of melanocyte-associated immunohistochemical markers in acral lentiginous melanoma and acral benign nevi. Am. J. Dermatopathol. 2023, 45, 748–752. [Google Scholar] [CrossRef]
  16. Rasic, D.; Korsgaard, N.; Marcussen, N.; Precht Jensen, E.M. Diagnostic utility of combining PRAME and HMB-45 stains in primary melanocytic tumors. Ann. Diagn. Pathol. 2023, 67, 152211. [Google Scholar] [CrossRef] [PubMed]
  17. Bahmad, H.F.; Oh, K.S.; Alexis, J. Potential diagnostic utility of PRAME and p16 immunohistochemistry in melanocytic nevi and malignant melanoma. J. Cutan. Pathol. 2023, 50, 763–772. [Google Scholar] [CrossRef] [PubMed]
  18. Kunc, M.; Żemierowska, N.; Skowronek, F.; Biernat, W. Diagnostic test accuracy meta-analysis of PRAME in distinguishing primary cutaneous melanomas from benign melanocytic lesions. Histopathology 2023, 83, 3–14. [Google Scholar] [CrossRef] [PubMed]
  19. Ricci, C.; Altavilla, M.V.; Corti, B.; Pasquini, E.; Presutti, L.; Baietti, A.M.; Amorosa, L.; Balbi, T.; Baldovini, C.; Ambrosi, F.; et al. PRAME expression in mucosal melanoma of the head and neck region. Am. J. Surg. Pathol. 2023, 47, 599–610. [Google Scholar] [CrossRef] [PubMed]
  20. Miao, Q.J.; Zang, J.; Shao, X.B.; Sun, J.F.; Chen, Y.P.; Chen, H. Analysis of PRAME immunocytochemistry in 109 acral malignant melanoma in situ. J. Clin. Pathol. 2023; online ahead of print. [Google Scholar] [CrossRef]
  21. Cazzato, G.; Cascardi, E.; Colagrande, A.; Belsito, V.; Lospalluti, L.; Foti, C.; Arezzo, F.; Dellino, M.; Casatta, N.; Lupo, C.; et al. PRAME Immunoexpression in 275 Cutaneous Melanocytic Lesions: A Double Institutional Experience. Diagnostics 2022, 12, 2197. [Google Scholar] [CrossRef]
  22. Koh, S.S.; Lau, S.K.; Scapa, J.V. Cassarino DS. PRAME immunohistochemistry of spitzoid neoplasms. J. Cutan. Pathol. 2022, 49, 709–716. [Google Scholar] [CrossRef]
  23. Ronchi, A.; Zito Marino, F.; Moscarella, E.; Brancaccio, G.; Argenziano, G.; Troiani, T.; Napolitano, S.; Franco, R.; Cozzolino, I. PRAME Immunocytochemistry for the diagnosis of melanoma metastases in cytological samples. Diagnostics 2022, 12, 646. [Google Scholar] [CrossRef] [PubMed]
  24. Lezcano, C.; Jungbluth, A.A.; Nehal, K.S.; Hollmann, T.J.; Busam, K.J. PRAME expression in melanocytic tumors. Am. J. Surg. Pathol. 2018, 42, 1456–1465. [Google Scholar] [CrossRef] [PubMed]
  25. Lezcano, C.; Müller, A.M.; Frosina, D.; Hernandez, E.; Geronimo, J.A.; Busam, K.J.; Jungbluth, A.A. Immunohistochemical detection of cancer-testis antigen PRAME. Int. J. Surg. Pathol. 2021, 29, 826–835. [Google Scholar] [CrossRef] [PubMed]
  26. Iura, K.; Kohashi, K.; Hotokebuchi, Y.; Ishii, T.; Maekawa, A.; Yamada, Y.; Yamamoto, H.; Iwamoto, Y.; Oda, Y. Cancer-testis antigens PRAME and NY-ESO-1 correlate with tumour grade and poor prognosis in myxoid liposarcoma. J. Pathol. Clin. Res. 2015, 1, 144–159. [Google Scholar] [CrossRef] [PubMed]
  27. Iura, K.; Maekawa, A.; Kohashi, K.; Ishii, T.; Bekki, H.; Otsuka, H.; Yamada, Y.; Yamamoto, H.; Harimaya, K.; Iwamoto, Y.; et al. Cancer-testis antigen expression in synovial sarcoma: NY-ESO-1, PRAME, MAGEA4, and MAGEA1. Hum. Pathol. 2017, 61, 130–139. [Google Scholar] [CrossRef]
  28. Oberthuer, A.; Hero, B.; Spitz, R.; Berthold, F.; Fischer, M. The tumor-associated antigen PRAME is universally expressed in high-stage neuroblastoma and associated with poor outcome. Clin. Cancer Res. 2004, 10, 4307–4313. [Google Scholar] [CrossRef] [PubMed]
  29. Neumann, E.; Engelsberg, A.; Decker, J.; Störkel, S.; Jaeger, E.; Huber, C.; Seliger, B. Heterogeneous expression of the tumor-associated antigens RAGE-1, PRAME, and glycoprotein 75 in human renal cell carcinoma: Candidates for T-cell-based immunotherapies? Cancer Res. 1998, 58, 4090–4095. [Google Scholar]
  30. Taniguchi, Y.; Ishida, M.; Saito, T.; Ryota, H.; Utsumi, T.; Maru, N.; Matsui, H.; Hino, H.; Tsuta, K.; Murakawa, T. Preferentially expressed antigen in melanoma as a novel diagnostic marker differentiating thymic squamous cell carcinoma from thymoma. Sci. Rep. 2020, 10, 12286. [Google Scholar] [CrossRef]
  31. Hodgson, A.; Jungbluth, A.A.; Katabi, N.; Xu, B.; Downes, M.R. Evaluation of cancer testis antigen (CT10, PRAME) and MHC I expression in high-grade urothelial carcinoma of the bladder. Virchows Arch. 2020, 476, 535–542. [Google Scholar] [CrossRef]
  32. Dyrskjøt, L.; Zieger, K.; Kissow Lildal, T.; Reinert, T.; Gruselle, O.; Coche, T.; Borre, M.; Ørntoft, T.F. Expression of MAGE-A3, NY-ESO-1, LAGE-1 and PRAME in urothelial carcinoma. Br. J. Cancer 2012, 107, 116–122. [Google Scholar] [CrossRef]
  33. Gezgin, G.; Luk, S.J.; Cao, J.; Dogrusöz, M.; van der Steen, D.M.; Hagedoorn, R.S.; Krijgsman, D.; van der Velden, P.A.; Field, M.G.; Luyten, G.P.M.; et al. PRAME as a potential target for immunotherapy in metastatic uveal melanoma. JAMA Ophthalmol. 2017, 135, 541–549. [Google Scholar] [CrossRef] [PubMed]
  34. Pujol, J.L.; De Pas, T.; Rittmeyer, A.; Vallières, E.; Kubisa, B.; Levchenko, E.; Wiesemann, S.; Masters, G.A.; Shen, R.; Tjulandin, S.A.; et al. Safety and immunogenicity of the PRAME cancer immunotherapeutic in patients with resected non-small cell lung cancer: A Phase I dose escalation study. J. Thorac. Oncol. 2016, 11, 2208–2217. [Google Scholar] [CrossRef]
  35. Nguyen, J.K.; Przybycin, C.G.; McKenney, J.K.; Magi-Galluzzi, C. Immunohistochemical staining patterns of Ki-67 and p53 in florid reactive urothelial atypia and urothelial carcinoma in situ demonstrate significant overlap. Hum. Pathol. 2020, 98, 81–88. [Google Scholar] [CrossRef] [PubMed]
  36. Cammareri, C.; Beltzung, F.; Michal, M.; Vanhersecke, L.; Coindre, J.M.; Velasco, V.; Le Loarer, F.; Vergier, B.; Perret, R. PRAME immunohistochemistry in soft tissue tumors and mimics: A study of 350 cases highlighting its imperfect specificity but potentially useful diagnostic applications. Virchows Arch. 2023, 483, 145–156. [Google Scholar] [CrossRef] [PubMed]
  37. Ricci, C.; Franceschini, T.; Giunchi, F.; Grillini, M.; Ambrosi, F.; Massari, F.; Mollica, V.; Colecchia, M.; Fiorentino, M. Immunohistochemical expression of preferentially expressed antigen in melanoma (PRAME) in the uninvolved background testis, germ cell neoplasia in situ, and germ cell tumors of the testis. Am. J. Clin. Pathol. 2022, 157, 644–648. [Google Scholar] [CrossRef]
  38. Mitsuhashi, K.; Masuda, A.; Wang, Y.H.; Shiseki, M.; Motoji, T. Prognostic significance of PRAME expression based on immunohistochemistry for diffuse large B-cell lymphoma patients treated with R-CHOP therapy. Int. J. Hematol. 2014, 100, 88–95. [Google Scholar] [CrossRef] [PubMed]
  39. Le, M.K.; Vuong, H.G.; Dunn, I.F.; Kondo, T. Molecular and clinicopathological implications of PRAME expression in adult glioma. PLoS ONE 2023, 18, e0290542. [Google Scholar] [CrossRef]
  40. Pan, S.H.; Su, K.Y.; Spiessens, B.; Kusuma, N.; Delahaye, N.F.; Gruselle, O.; Myo, A.; de Creus, A.; Louahed, J.; Chang, G.C.; et al. Gene expression of MAGE-A3 and PRAME tumor antigens and EGFR mutational status in Taiwanese non-small cell lung cancer patients. Asia Pac. J. Clin. Oncol. 2017, 13, e212–e223. [Google Scholar] [CrossRef]
  41. Thongprasert, S.; Yang, P.C.; Lee, J.S.; Soo, R.; Gruselle, O.; Myo, A.; Louahed, J.; Lehmann, F.F.; Brichard, V.G.; Coche, T. The prevalence of expression of MAGE-A3 and PRAME tumor antigens in East and South East Asian non-small cell lung cancer patients. Lung Cancer 2016, 101, 137–144. [Google Scholar] [CrossRef]
  42. See, S.H.C.; Smith, S.H.; Finkelman, B.S.; LaBoy, C.; Novo, J.E.; Siziopikou, K.P.; Blanco, L.Z., Jr. The role of PRAME and NY-ESO-1 as potential therapeutic and prognostic biomarkers in triple-negative breast carcinomas. Pathol. Res. Pract. 2023, 241, 154299. [Google Scholar] [CrossRef]
  43. Tessari, A.; Pilla, L.; Silvia, D.; Duca, M.; Paolini, B.; Carcangiu, M.L.; Mariani, L.; de Braud, F.G.; Cresta, S. Expression of NY-ESO-1, MAGE-A3, PRAME and WT1 in different subgroups of breast cancer: An indication to immunotherapy? Breast 2018, 42, 68–73. [Google Scholar] [CrossRef] [PubMed]
  44. Doolan, P.; Clynes, M.; Kennedy, S.; Mehta, J.P.; Crown, J.; O’Driscoll, L. Prevalence and prognostic and predictive relevance of PRAME in breast cancer. Breast Cancer Res. Treat. 2008, 109, 359–365. [Google Scholar] [CrossRef] [PubMed]
  45. Vlasenkova, R.; Konysheva, D.; Nurgalieva, A.; Kiyamova, R. Characterization of Cancer/Testis Antigens as Prognostic Markers of Ovarian Cancer. Diagnostics 2023, 13, 3092. [Google Scholar] [CrossRef]
  46. Šafanda, A.; KendallBártů, M.; Michálková, R.; Stružinská, I.; Drozenová, J.; Fabián, P.; Hausnerová, J.; Laco, J.; Matěj, R.; Škapa, P.; et al. Immunohistochemical expression of PRAME in 485 cases of epithelial tubo-ovarian tumors. Virchows Arch. 2023, 483, 509–516. [Google Scholar] [CrossRef] [PubMed]
  47. Ogul, A.; Paydas, S.; Doran, F.; Erdogan, K.E.; Tohumcuoglu, M.; Buyuksimsek, M.; Mirili, C.; Yetisir, A.E. The effect of preferentially expressed antigen in melanoma (PRAME) expression status on survival in stage II and stage III colon cancer. J. BUON 2021, 26, 992–1001. [Google Scholar]
  48. Oyama, K.; Kanki, K.; Shimizu, H.; Kono, Y.; Azumi, J.; Toriguchi, K.; Hatano, E.; Shiota, G. Impact of preferentially expressed antigen of melanoma on the prognosis of hepatocellular carcinoma. Gastrointest. Tumors 2017, 3, 128–135. [Google Scholar] [CrossRef]
  49. Lee, Y.K.; Park, U.H.; Kim, E.J.; Hwang, J.T.; Jeong, J.C.; Um, S.J. Tumor antigen PRAME is up-regulated by MZF1 in cooperation with DNA hypomethylation in melanoma cells. Cancer Lett. 2017, 403, 144–151. [Google Scholar] [CrossRef]
  50. Nettersheim, D.; Arndt, I.; Sharma, R.; Riesenberg, S.; Jostes, S.; Schneider, S.; Hölzel, M.; Kristiansen, G.; Schorle, H. The cancer/testis-antigen PRAME supports the pluripotency network and represses somatic and germ cell differentiation programs in seminomas. Br. J. Cancer 2016, 115, 454–464. [Google Scholar] [CrossRef]
  51. Tajeddine, N.; Louis, M.; Vermylen, C.; Gala, J.L.; Tombal, B.; Gailly, P. Tumor associated antigen PRAME is a marker of favorable prognosis in childhood acute myeloid leukemia patients and modifies the expression of S100A4, Hsp 27, p21, IL-8 and IGFBP-2 in vitro and in vivo. Leuk. Lymphoma 2008, 49, 1123–1131. [Google Scholar] [CrossRef]
  52. Tanaka, N.; Wang, Y.H.; Shiseki, M.; Takanashi, M.; Motoji, T. Inhibition of PRAME expression causes cell cycle arrest and apoptosis in leukemic cells. Leuk. Res. 2011, 35, 1219–1225. [Google Scholar] [CrossRef]
  53. Zhang, W.; Li, L.; Cai, L.; Liang, Y.; Xu, J.; Liu, Y.; Zhou, L.; Ding, C.; Zhang, Y.; Zhao, H.; et al. Tumor-associated antigen Prame targets tumor suppressor p14/ARF for degradation as the receptor protein of CRL2Prame complex. Cell Death Differ. 2021, 28, 1926–1940. [Google Scholar] [CrossRef] [PubMed]
  54. Neal, D.J.; Amin, M.B.; Smith, S.C. CK20 versus AMACR and p53 immunostains in evaluation of urothelial carcinoma in situ and Reactive Atypia. Diagn. Pathol. 2020, 15, 61. [Google Scholar] [CrossRef] [PubMed]
  55. Wymer, K.M.; Sharma, V.; Saigal, C.S.; Chamie, K.; Litwin, M.S.; Packiam, V.T.; Mossanen, M.; Pagliaro, L.C.; Borah, B.J.; Boorjian, S.A. Cost-effectiveness analysis of pembrolizumab for Bacillus Calmette-Guérin-unresponsive carcinoma in situ of the bladder. J. Urol. 2021, 205, 1326–1335. [Google Scholar] [CrossRef] [PubMed]
  56. Balar, A.V.; Kamat, A.M.; Kulkarni, G.S.; Uchio, E.M.; Boormans, J.L.; Roumiguié, M.; Krieger, L.E.M.; Singer, E.A.; Bajorin, D.F.; Grivas, P.; et al. Pembrolizumab monotherapy for the treatment of high-risk non-muscle-invasive bladder cancer unresponsive to BCG (KEYNOTE-057): An open-label, single-arm, multicentre, phase 2 study. Lancet Oncol. 2021, 22, 919–930. [Google Scholar] [CrossRef]
  57. A Boorjian, S.; Alemozaffar, M.; Konety, B.R.; Shore, N.D.; Gomella, L.G.; Kamat, A.M.; Bivalacqua, T.J.; Montgomery, J.S.; Lerner, S.P.; E Busby, J.; et al. Intravesical nadofaragene firadenovec gene therapy for BCG-unresponsive non-muscle-invasive bladder cancer: A single-arm, open-label, repeat-dose clinical trial. Lancet Oncol. 2021, 22, 107–117. [Google Scholar] [CrossRef]
  58. Brohl, A.S.; Sindiri, S.; Wei, J.S.; Milewski, D.; Chou, H.-C.; Song, Y.K.; Wen, X.; Kumar, J.; Reardon, H.V.; Mudunuri, U.S.; et al. Immuno-transcriptomic profiling of extracranial pediatric solid malignancies. Cell Rep. 2021, 37, 110047. [Google Scholar] [CrossRef] [PubMed]
  59. Kewitz, S.; Staege, M.S. Knock-down of PRAME increases retinoic acid signaling and cytotoxic drug sensitivity of Hodgkin lymphoma cells. PLoS ONE 2013, 8, e55897. [Google Scholar] [CrossRef]
Figure 1. Typical histopathological features of carcinoma in situ of the urinary tract. Non-papillary flat neoplastic growth of the neoplastic cells having large round to oval nuclei (hematoxylin and eosin, ×400).
Figure 1. Typical histopathological features of carcinoma in situ of the urinary tract. Non-papillary flat neoplastic growth of the neoplastic cells having large round to oval nuclei (hematoxylin and eosin, ×400).
Diagnostics 13 03636 g001
Figure 2. Immunohistochemical staining for PRAME. PRAME is expressed in the nuclei of the neoplastic cells of carcinoma in situ of the urinary tract (×200).
Figure 2. Immunohistochemical staining for PRAME. PRAME is expressed in the nuclei of the neoplastic cells of carcinoma in situ of the urinary tract (×200).
Diagnostics 13 03636 g002
Figure 3. Immunohistochemical stainings for p53. (a) Diffuse expression pattern. (b) Null expression pattern. (c) Wild-type expression pattern (×200, each).
Figure 3. Immunohistochemical stainings for p53. (a) Diffuse expression pattern. (b) Null expression pattern. (c) Wild-type expression pattern (×200, each).
Diagnostics 13 03636 g003aDiagnostics 13 03636 g003b
Table 1. Correlation between PRAME expression and p53 expression patterns in carcinoma in situ of the urinary tract.
Table 1. Correlation between PRAME expression and p53 expression patterns in carcinoma in situ of the urinary tract.
p53 Aberrant Patternp53 Wild Pattern
PRAME positive1310
PRAME negative1614
PRAME, preferentially expressed antigen in melanoma.
Table 2. Summary of PRAME expression in urothelial carcinoma.
Table 2. Summary of PRAME expression in urothelial carcinoma.
PRAME ExpressionDetection MethodsReferences
7% (histological grade not available)IHC[13]
15% of muscle-invasive high-grade UCIHC[31]
3% of non-invasive papillary UC
16% of pT1 UC
28% of pT2-4 UC
RT-PCR[32]
43.4% of CISIHCPresent study
CIS, carcinoma in situ; IHC, immunohistochemistry; PRAME, preferentially expressed antigen in melanoma; UC, urothelial carcinoma.
Table 3. Summary of PRAME expression in various types of malignant tumor.
Table 3. Summary of PRAME expression in various types of malignant tumor.
Histological Type of TumorFrequency of PRAME ExpressionReferences
Non-epithelial tumors
Malignant melanoma
SkinHigh[13,14,15,16,17,18,20,21,22,23,24]
MucosalHigh[19,25,33]
Myxoid liposarcomaHigh[13,25,26,36]
Synovial sarcomaHigh[13,25,27,36]
NeuroblastomaHigh[13,25,28]
SeminomaIntermediate–high[13,25,37]
RhabdomyosarcomaIntermediate[13,25,36]
Malignant peripheral nerve sheath tumorLow–intermediate[13,25,36]
AngiosarcomaLow–intermediate[13,25,36]
Malignant lymphomaLow–intermediate[13,38]
Ewing sarcomaLow[13,25,36]
LeiomyosarcomaLow[13,25,36]
GlioblastomaLow[13,39]
OsteosarcomaLow[13]
Epithelial tumors
Thymic cancerHigh[13,30]
Lung cancerIntermediate–high[13,25,40,41]
Breast cancerIntermediate–high[13,25,42,43,44]
Ovarian cancerIntermediate[13,25,45,46]
Renal cell carcinomaLow–intermediate[13,25,29]
Colorectal cancerLow–intermediate[13,47]
Hepatocellular carcinomaLow[13,48]
Pancreatic cancerLow[13]
PRAME, preferentially expressed antigen in melanoma.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fujii, S.; Ishida, M.; Komura, K.; Nishimura, K.; Tsujino, T.; Saito, T.; Taniguchi, Y.; Murakawa, T.; Azuma, H.; Hirose, Y. Expression of Preferentially Expressed Antigen in Melanoma, a Cancer/Testis Antigen, in Carcinoma In Situ of the Urinary Tract. Diagnostics 2023, 13, 3636. https://doi.org/10.3390/diagnostics13243636

AMA Style

Fujii S, Ishida M, Komura K, Nishimura K, Tsujino T, Saito T, Taniguchi Y, Murakawa T, Azuma H, Hirose Y. Expression of Preferentially Expressed Antigen in Melanoma, a Cancer/Testis Antigen, in Carcinoma In Situ of the Urinary Tract. Diagnostics. 2023; 13(24):3636. https://doi.org/10.3390/diagnostics13243636

Chicago/Turabian Style

Fujii, Shota, Mitsuaki Ishida, Kazumasa Komura, Kazuki Nishimura, Takuya Tsujino, Tomohito Saito, Yohei Taniguchi, Tomohiro Murakawa, Haruhito Azuma, and Yoshinobu Hirose. 2023. "Expression of Preferentially Expressed Antigen in Melanoma, a Cancer/Testis Antigen, in Carcinoma In Situ of the Urinary Tract" Diagnostics 13, no. 24: 3636. https://doi.org/10.3390/diagnostics13243636

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop