Preferentially Expressed Antigen in Melanoma and Malignant Melanoma: Comparison
Please note this is a comparison between Version 2 by Lindsay Dong and Version 1 by Gerardo Cazzato.

Preferentially expressed antigen in melanoma (PRAME) is a cancer testis antigen (CTA) identified in 1997 through analysis of the specificity of tumor-reactive T-cell clones derived from a patient with metastatic cutaneous melanoma. Although at first it seemed even more specific, various studies have shown that PRAME can also be expressed in the context of atypical lesions that do not correspond solely to the definition of malignant melanoma. Various studies have shown that PRAME can also be expressed in the context of atypical lesions apart from MM and, for this reason, the diagnostic sensitivity and specificity (hence accuracy) are clearly lower. 

  • PRAME
  • malignant melanoma
  • skin

1. Introduction

Despite advances in therapy and treatment, malignant melanoma (MM) continues to be a very aggressive skin cancer, with 324,635 new cases and 57,043 deaths worldwide in 2020, accounting for 1.7% of all cancers in the world population [1], and showing a rising incidence [2]. Histological diagnosis continues to be of great importance in the diagnostic and therapeutic care pathway of patients suffering from malignant melanoma [3] and although it is often relatively simple, at other times, the recognition of the malignant lesion can pose a great challenge [4]. For this reason, alongside conventional histopathology, ancillary immunohistochemistry techniques have been introduced, in an attempt to simplify the difficulties of diagnosis [5]. Despite this, researchers have always tried to find new markers that could help, particularly in challenging cases of MM [6], such as Melan-A (MART-1) [7], HMB-45 (anti-human melanosome clone HMB45) [8], and MITF (melanocyte inducing transcription factor) [9], but the morphological difficulty together with the negativity of some histotypes of MM (such as desmoplastic melanoma) [10], has meant that even these have failed to completely resolve the diagnostic dilemmas. The widespread use of Sry-related HMg-Box gene 10 (SOX-10) has made it possible to solve cases of differential diagnostics (such as differential diagnoses between some histotypes of MM and other types of skin lesions) [11[11][12],12], but a certain degree of difficulty still remains. PRAME (preferentially expressed antigen in melanoma) is a cancer testis antigen (CTA) that was first identified in 1997 through analysis of the specificity of tumor-reactive T-cell clones derived from a patient with metastatic cutaneous melanoma [13]. Over the years, however, it was found that PRAME was not only expressed in cutaneous melanoma, but also in other types of MM including ocular, as well as in various malignant neoplasms not of melanocyte origin, such as non-small cell lung cancer [14], breast cancer [15], renal cancer [16], ovarian cancer [17], hematological malignancies [18[18][19][20],19,20], synovial sarcoma, and myxoid liposarcoma [21,22][21][22]. Healthy tissues do not express PRAME except the testis, ovary, placenta, adrenal glands, and endometrium [23].

2. Preferentially Expressed Antigen in Melanoma and Malignant Melanoma

Since its first description back in 1997 by Ideka et al. [13], PRAME has been of interest both as a possible diagnostic parameter and as a target for possible immunotherapy. Since then, various studies have tried to shed light on the real potential use of this marker, and in recent years the aspect relating to immunoexpression has been particularly studied in the context of differential histopathological diagnostics of MM [25,26,27][24][25][26]. Recent works by Lezcano C. et al. [25,26,28][24][25][27] have clarified some important aspects relating to the role of PRAME as a diagnostic aid. In fact, in one of the first studies conducted on this topic, the autscholars [25][24] presented their findings regarding the immunoexpression of PRAME in 400 melanocytic tumors, including 155 primary and 100 metastatic melanomas, and 145 melanocytic nevi. Diffuse nuclear immunoreactivity for PRAME was found in 87% of metastatic and 83.2% of primary melanomas. Among melanoma subtypes, PRAME was diffusely expressed in 94.4% of acral melanomas, 92.5% of superficial spreading melanomas, 90% of nodular melanomas, 88.6% of lentigo maligna melanomas, and 35% of desmoplastic melanomas. When in situ and nondesmoplastic invasive melanoma components were present, PRAME expression was seen in both. Of the 140 cutaneous melanocytic nevi, 86.4% were completely negative for PRAME. Immunoreactivity for PRAME was seen, albeit usually only in a minor subpopulation of lesional melanocytes, in 13.6% of cutaneous nevi, including dysplastic nevi, common acquired nevi, traumatized and recurrent nevi, and Spitz nevi (one case in a 6 years-old child). Rare isolated junctional melanocytes with immunoreactivity for PRAME were also seen in solar lentigines and benign non lesional skin. In this first work, it was highlighted that this marker could certainly constitute a valid support in the differential diagnosis of benign and malignant pigmented lesions, albeit with limitations in selected cases. In a 2020 paper, Lezcano et al. [26,28][25][27] reported a series of cases of immunostaining for PRAME: they examined 45 nodal melanocytic deposits comprising 30 nodal nevi and 15 melanoma metastases. All nodal nevi (30/30) were negative for PRAME, whereas all melanoma metastases (15/15) were diffusely positive for PRAME IHC. Furthermore, the autscholars reported the utility of PRAME/Melan A dual-label immunostaining and stressed the usefulness of PRAME IHC in the assessment of diagnostically challenging nodal melanocytic deposits, such as intraparenchymal nodal nevi, metastases confined to the capsular fibrous tissue, or in the setting of small metastases coexisting with a nodal nevus in the same lymph node. These aspects were also analyzed by See et al. [29][28]. In a paper published in September 2020, Gradecki S. et al. reported their experience of immunostaining of PRAME on 155 cases of metastatic melanoma, 54 of which were to lymph node and 101 to non-lymph node sites. PRAME expression was seen in 151/155 (97.4%) cases, with 4+ expression in 64 cases (41.3%), 3+ expression in 46 cases (29.7%), 2+ expression in 18 cases (11.6%), and 1+ expression in 23 cases (14.8%). Lymph node metastases were more likely to show a lower expression as compared to metastases to other anatomic sites. Based on these data, the authoscholars suggested the possibility that PRAME could be of diagnostic aid in confirming a diagnosis of MM in a metastatic setting (both lymph node and other sites) [30][29].

Considering that in the biology of malignant transformation from dysplastic nevus to MM a series of genetic-molecular events occur, that alter the expression of various protein molecules [31[30][31],32], Lohman et al. [33][32] investigated the immunoexpression pattern of PRAME in melanomas associated with a dysplastic nevus (NAM). In tThat paper they reviewedere are thirty-six cases: 67% (24/36) of melanomas were PRAME positive (4+) while no (0/36) nevi showed 4+ positivity; 81% (29/36) of nevi were completely PRAME negative compared to 17% (6/36) of melanomas. In 67% of cases (24/36) PRAME differentiated between benign and malignant melanocyte populations. The authors identified a high rate (67%) of differential PRAME staining in adjacent benign and malignant melanocyte populations in NAM. In PRAME positive (4+) melanomas, PRAME differentiates 100% (24/24) of benign and malignant melanocyte populations. When 4+ staining is used as the threshold for positivity, PRAME staining has a sensitivity of 67% (24/36) and a specificity of 100% (36/36). These results supported the concept that PRAME IHC can assist in distinguishing melanocyte populations in melanoma arising within nevi.

A study by Raghavan et al. investigated further and confirmed the usefulness of PRAME IHC for making differential diagnoses between melanocytic proliferations with intermediate histopathology and melanoma: they found that traumatized, mitotically-active, persistent and recurrent, and dysplastic nevi usually lacked PRAME expression altogether. As in the study by Lezcano et al. [26][25], the need for further histopathological, cytogenetic, and molecular characteristics to interpret the PRAME status in cases of spitzoid neoplasia was confirmed. Indeed, although most benign and intermediate Spitz lesions lacked widespread PRAME expression, widespread PRAME positivity was observed in a Spitz nevus and atypical Spitz tumor. Furthermore, immunoreactivity intensity ranged from weak to strong, regardless of the degree of atypia [34][33].

PRAME expression was associated with the largest basal diameter (LBD) and tumor volume. Notwithstanding, PRAME expression can appear at any stage of tumor progression and not only in advanced stages. These findings were in agreement with the already known strong association of LBD with GEP and metastatic risk [35][34].

Very recently, in 2022, Krajisnik et al. published a paper where they evaluated the expression of the PRAME protein in a series of melanocytic lesions of the nail. In their work, 25 Twenty-five nail unit melanomas (including small biopsy and amputation samples) and 32 control benign melanocytic lesions were retrospectively reviewed. PRAME nuclear staining was evaluated (similarly to other works in the literature) as a percentage and intensity labeling. All melanoma cases showed PRAME nuclear expression, which was usually widespread and strong. In samples with few cancer cells, staining was restricted to tumor cells, corresponding to the initial H&E imprint. All control cases were negative for PRAME expression. The expression of PRAME was useful in distinguishing between melanomas and other melanocytic lesions of the nail. This antibody has also been shown to be diagnostically valuable for detecting melanoma cells in small samples with minimal disease [62][35].

 3. Summary

PRAME expression in melanoma cells is regulated by many actors. Hypermethylation appears to downregulate the expression of PRAME [63][36], while downregulation of miR-211 (an RNA gene) results in increased expression [64][37]. Aberrant PRAME hypomethylation results in an augmented transcription and a higher risk of metastasis in uveal melanoma. [65][38] The activity of MZF-1 also increases the expression of PRAME in melanoma cells and their ability to form colonies [63,66][36][39]. A retrospective study conducted on different types of mucosal melanoma (sinonasal, gastrointestinal, genitourinary, and oropharyngeal) shows the prognostic implication of PRAME expression. PRAME expression was lower in surviving patients at 24 months, and higher PRAME staining detected on immunohistochemical analysis was correlated with a 170% risk of death [67][40]. Furthermore, mucosal melanomas analyzed by Toyama et al. [67][40] showed a higher PRAME expression in cells with an epithelioid than a spindle morphology.
From a molecular point of view, mucosal melanomas are more similar to uveal melanoma than to cutaneous melanoma [25,46,68][24][41][42].
PRAME is expressed by melanoma and many different tumors: a higher expression is typical of advanced diseases and is associated with lymph node spread of the disease. Prognosis is poor when PRAME is overexpressed, indicating worse disease-free, progression-free, and metastasis-free prognosis, and worse overall survival [69][43].

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. Sandru, A.; Voinea, S.; Panaitescu, E.; Blidaru, A. Survival rates of patients with metastatic malignant melanoma. J. Med. Life 2014, 7, 572–576.
  3. Cabrera, R.; Recule, F. Unusual Clinical Presentations of Malignant Melanoma: A Review of Clinical and Histologic Features with Special Emphasis on Dermatoscopic Findings. Am. J. Clin. Dermatol. 2018, 19 (Suppl. 1), 15–23.
  4. Bsirini, C.; Smoller, B.R. Histologic mimics of malignant melanoma. Singapore Med. J. 2018, 59, 602–607.
  5. Luca, R. Diagnostic, Prognostic and Predictive Immunohistochemistry in Malignant Melanoma of the Skin. Klin. Onkol. 2018, 31, 152–155.
  6. Barnhill, L.R. Pathology of Malignant Melanoma, 1st ed.; Springer: Milan, Italy, 2004; pp. 33–39.
  7. Kim, J.; Taube, J.M.; McCalmont, T.H.; Glusac, E.J. Quantitative comparison of MiTF, Melan-A, HMB-45 and Mel-5 in solar lentigines and melanoma in situ. J. Cutan. Pathol. 2011, 38, 775–779.
  8. Gleason, B.C.; Nascimento, A.F. HMB-45 and Melan-A are useful in the differential diagnosis between granular cell tumor and malignant melanoma. Am. J. Dermatopathol. 2007, 29, 22–27.
  9. Torres-Cabala, C.; Li-Ning-Tapia, E.; Hwu, W.J. Pathology-based Biomarkers Useful for Clinical Decisions in Melanoma. Arch. Med. Res. 2020, 51, 827–838.
  10. Kooper-Johnson, S.; Mahalingam, M.; Loo, D.S. SOX-10 and S100 Negative Desmoplastic Melanoma: Apropos a Diagnostically Challenging Case. Am. J. Dermatopathol. 2020, 42, 697–699.
  11. Plaza, J.A.; Bonneau, P.; Prieto, V.; Sangueza, M.; Mackinnon, A.; Suster, D.; Bacchi, C.; Estrozi, B.; Kazakov, D.; Kacerovska, D.; et al. Desmoplastic melanoma: An updated immunohistochemical analysis of 40 cases with a proposal for an additional panel of stains for diagnosis. J. Cutan. Pathol. 2016, 43, 313–323.
  12. Willis, B.C.; Johnson, G.; Wang, J.; Cohen, C. SOX10: A useful marker for identifying metastatic melanoma in sentinel lymph nodes. Appl. Immunohistochem. Mol. Morphol. 2015, 23, 109–112.
  13. 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.
  14. 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.
  15. Sun, Z.; Wu, Z.; Zhang, F.; Guo, Q.; Li, L.; Li, K.; Chen, H.; Zhao, J.; Song, D.; Huang, Q.; et al. PRAME is critical for breast cancer growth and metastasis. Gene 2016, 594, 160–164.
  16. 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.
  17. Zhang, W.; Barger, C.J.; Eng, K.H.; Klinkebiel, D.; Link, P.A.; Omilian, A.; Bshara, W.; Odunsi, K.; Karpf, A.R. PRAME expression and promoter hypomethylation in epithelial ovarian cancer. Oncotarget 2016, 7, 45352–45369.
  18. Matsushita, M.; Yamazaki, R.; Ikeda, H.; Kawakami, Y. Preferentially expressed antigen of melanoma (PRAME) in the development of diagnostic and therapeutic methods for hematological malignancies. Leuk. Lymphoma 2003, 44, 439–444.
  19. Oehler, V.G.; Guthrie, K.A.; Cummings, C.L.; Sabo, K.; Wood, B.L.; Gooley, T.; Yang, T.; Epping, M.T.; Shou, Y.; Pogosova-Agadjanyan, E.; et al. The preferentially expressed antigen in melanoma (PRAME) inhibits myeloid differentiation in normal hematopoietic and leukemic progenitor cells. Blood 2009, 114, 3299–3308.
  20. Quintarelli, C.; Dotti, G.; De Angelis, B.; Hoyos, V.; Mims, M.; Luciano, L.; Heslop, H.E.; Rooney, C.M.; Pane, F.; Savoldo, B. Cytotoxic T lymphocytes directed to the preferentially expressed antigen of melanoma (PRAME) target chronic myeloid leukemia. Blood 2008, 112, 1876–1885.
  21. Albertsmeier, M.; Altendorf-Hofmann, A.; Lindner, L.H.; Issels, R.D.; Kampmann, E.; Dürr, H.R.; Schubert-Fritschle, G.; Angele, M.K.; Kirchner, T.; Jungbluth, A.A.; et al. Cancer Testis Antigens and Immunotherapy: Expression of PRAME Is Associated with Prognosis in Soft Tissue Sarcoma. Cancers 2020, 12, 3612.
  22. Luk, S.J.; van der Steen, D.M.; Hagedoorn, R.S.; Jordanova, E.S.; Schilham, M.W.; Bovée, J.V.; Cleven, A.H.; Falkenburg, J.F.; Szuhai, K.; Heemskerk, M.H. PRAME and HLA Class I expression patterns make synovial sarcoma a suitable target for PRAME specific T-cell receptor gene therapy. Oncoimmunology 2018, 7, e1507600.
  23. Al-Khadairi, G.; Decock, J. Cancer Testis Antigens and Immunotherapy: Where Do We Stand in the Targeting of PRAME? Cancers 2019, 11, 984.
  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.
  25. Lezcano, C.; Pulitzer, M.; Moy, A.P.; Hollmann, T.J.; Jungbluth, A.A.; Busam, K.J. Immunohistochemistry for PRAME in the Distinction of Nodal Nevi From Metastatic Melanoma. Am. J. Surg. Pathol. 2020, 44, 503–508.
  26. Schefler, A.C.; Kim, R.S. Recent advancements in the management of retinoblastoma and uveal melanoma. Fac. Rev. 2021, 10, 51.
  27. Lezcano, C.; Jungbluth, A.A.; Busam, K.J. PRAME Immunohistochemistry as an Ancillary Test for the Assessment of Melanocytic Lesions. Surg. Pathol. Clin. 2021, 14, 165–175.
  28. See, S.H.C.; Finkelman, B.S.; Yeldandi, A.V. The diagnostic utility of PRAME and p16 in distinguishing nodal nevi from nodal metastatic melanoma. Pathol. Res. Pract. 2020, 216, 153105.
  29. Gradecki, S.E.; Slingluff, C.L.; Gru, A.A. PRAME expression in 155 cases of metastatic melanoma. J. Cutan. Pathol. 2021, 48, 479–485.
  30. Ko, J.M.; Velez, N.F.; Tsao, H. Pathways to melanoma. Semin. Cutan. Med. Surg. 2010, 29, 210–217.
  31. Borden, E.S.; Adams, A.C.; Buetow, K.H.; Wilson, M.A.; Bauman, J.E.; Curiel-Lewandrowski, C.; Chow, H.S.; LaFleur, B.J.; Hastings, K.T. Shared Gene Expression and Immune Pathway Changes Associated with Progression from Nevi to Melanoma. Cancers 2021, 14, 3.
  32. Lohman, M.E.; Steen, A.J.; Grekin, R.C.; North, J.P. The utility of PRAME staining in identifying malignant transformation of melanocytic nevi. J. Cutan. Pathol. 2021, 48, 856–862.
  33. Raghavan, S.S.; Wang, J.Y.; Kwok, S.; Rieger, K.E.; Novoa, R.A.; Brown, R.A. PRAME expression in melanocytic proliferations with intermediate histopathologic or spitzoid features. J. Cutan. Pathol. 2020, 47, 1123–1131.
  34. Schefler, A.C.; Koca, E.; Bernicker, E.H.; Correa, Z.M. Relationship between clinical features, GEP class, and PRAME expression in uveal melanoma. Graefes Arch Clin. Exp. Ophthalmol. 2019, 257, 1541–1545.
  35. Krajisnik, A.; Gharavi, N.M.; Faries, M.B.; Balzer, B.L.; Frishberg, D.P.; Martelli, M.; Shon, W. Immunohistochemistry for Preferentially Expressed Antigen in Melanoma in the Differential Diagnosis of Melanocytic Lesions of the Nail Apparatus. Am. J. Dermatopathol. 2022, 2.
  36. 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.
  37. Sakurai, E.; Maesawa, C.; Shibazaki, M.; Yasuhira, S.; Oikawa, H.; Sato, M.; Tsunoda, K.; Ishikawa, Y.; Watanabe, A.; Takahashi, K.; et al. Downregulation of microRNA-211 is involved in expression of preferentially expressed antigen of melanoma in melanoma cells. Int. J. Oncol. 2011, 39, 665–672.
  38. Field, M.G.; Durante, M.A.; Decatur, C.L.; Tarlan, B.; Oelschlager, K.M.; Stone, J.F.; Kuznetsov, J.; Bowcock, A.M.; Kurtenbach, S.; Harbour, J.W. Epigenetic reprogramming and aberrant expression of PRAME are associated with increased metastatic risk in Class 1 and Class 2 uveal melanomas. Oncotarget 2016, 7, 59209–59219.
  39. 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.
  40. Toyama, A.; Siegel, L.; Nelson, A.C.; Najmuddin, M.; Bu, L.; LaRue, R.; Henzler, C.; Caicedo-Granados, E.; Giubellino, A.; Li, F. Analyses of molecular and histopathologic features and expression of PRAME by immunohistochemistry in mucosal melanomas. Mod. Pathol. 2019, 32, 1727–1733.
  41. Field, M.G.; Decatur, C.L.; Kurtenbach, S.; Gezgin, G.; van der Velden, P.A.; Jager, M.J.; Kozak, K.N.; Harbour, J.W. PRAME as an Independent Biomarker for Metastasis in Uveal Melanoma. Clin. Cancer Res. 2016, 22, 1234–1242.
  42. Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-genome landscapes of major melanoma subtypes. Nature 2017, 545, 175–180.
  43. Li, J.; Yin, J.; Zhong, J.; Yang, Z.; Tang, A.; Li, S. Clinicopathological and Prognostic Significance of PRAME Overexpression in Human Cancer: A Meta-Analysis. BioMed. Res. Int. 2020, 2020, 8828579.
More
Video Production Service