Chapter 35 / Immunogenic -Associated 619 35 Role of Melanoma-Associated Antigens

Rishab K. Gupta, Ana M. McElrath-Garza, and Donald L. Morton

CONTENTS INTRODUCTION EVIDENCE FOR IMMUNITY TO HUMAN MELANOMA HOST IMMUNE RESPONSE AND DEVELOPMENT OF MALIGNANT MELANOMA RECOGNITION OF MAA EXPRESSED BY HUMAN MELANOMA CELLS ROLE OF IMMUNOGENIC MAA IN MELANOMA PROGRESSION OR INHIBITION CONCLUSIONS AND PERSPECTIVES REFERENCES

Summary The dynamic interaction between malignant melanoma cells and the host’s immune system has a profound influence on the development and progression of disease. Numerous studies have documented the therapeutic potential of immunological manipulation in patients with melanoma, and many tumor- associated antigens have been studied as possible targets of melanoma-specific immunotherapy. This chapter outlines the most significant melanoma-associated antigens, highlighting those with the greatest potential for influencing future treatment modalities. Specific antigens defined by T- and B-cells are discussed, and how these antigens are recognized, processed, and manipulated by the host’s immune system is explained. The role of gangliosides in distinguishing different stages of differentia- tion is described, and a 90-kDa tumor-associated glycoprotein which has considerable promise as a marker of melanoma tumor burden is introduced. Finally, this chapter reviews technological advances in assays used to detect various melanoma-associated antigens. Key Words: Melanoma; melanoma-associated antigens; tumor-associated antigens; immuno- therapy; TA90.

INTRODUCTION The malignant transformation of to melanoma can be characterized by changes in cytology, morphology, proliferative index, biochemistry, and expres- sion. Activation of in transformed cells leads to overexpression and/or unique expression of molecules, such as tumor-associated antigens. Some of these molecules may be present in normal cells but sequestered from the immune system. In transformed

From: From Melanocytes to Melanoma: The Progression to Malignancy Edited by: V. J. Hearing and S. P. L. Leong © Humana Press Inc., Totowa, NJ

619 620 From Melanocytes to Melanoma cells, the antigens are expressed on nonsequestered sites. Tumor-associated antigens are considered immunogenic when the host recognizes them as nonself and generates an immune response. This chapter reviews the different melanoma-associated antigens (MAA) (e.g., melanoma antigen gene [MAGE]-1, NY-ESO-1, and TA90) and their roles in immune surveillance and the development/progression of melanoma. More than 40 yr ago, anecdotal reports of an increased incidence of melanoma regres- sion in immunosuppressed patients, such as those with congenital immunodeficiency disorders, suggested an important immune component in the development and control of disease (1–6). The more recent discovery of tissue-specific and common tumor- associated melanoma antigens further supports the key role for the immune system in the development and progression of melanoma. Additional evidence for immune modula- tion by MAA includes the prognostic significance of lymphocytes and other tumor-cell infiltrates (7–9), the delayed recurrence of rapidly progressive disease after successful treatment of the primary tumor (10–12), the presence of tumor cells in lymphatics, peripheral blood, and operative wounds without subsequent development of metastases (13–20), and the elimination of circulating tumor cells before the establishment of metastatic colonies (21). Ongoing investigations to identify clinically relevant target antigens and enhance the production of corresponding autoantibodies should lead to improvements in immunotherapy for melanoma.

EVIDENCE FOR IMMUNITY TO HUMAN MELANOMA Spontaneous regression of primary and metastatic melanoma, although rare, has been well documented (1–6). Published reports have correlated regression with minor viral and bacterial infections, fever of unknown origin, changes in hormonal balance, and congenital immunodeficiency disorders. Development of metastases that are histologi- cally and functionally identical to the primary tumor has been reported 10 to 20 yr after successful treatment of the primary tumor. Delayed recurrence suggests a clinically effective antitumor response during the disease-free interval. Although biological, physi- ological, and endocrine factors could contribute to spontaneous regression and long- term remission, clinical evidence implicates humoral and cellular immunological mechanisms (22,23).

HOST IMMUNE RESPONSE AND DEVELOPMENT OF MALIGNANT MELANOMA Because malignant melanoma is one of the most immunogenic solid tumors, it has served as a prototype for investigations of active immunotherapy against cancer (24–27). Determining the mechanisms of tumor escape from immunosurveillance is necessary to formulate effective immunotherapy against tumor cells (28). Despite the highly immu- nogenic nature of melanoma cells, as evidenced by the infiltration of tumor antigen- specific T-cells, most tumors are not completely destroyed by the host and can lead to progression of disease. An effective immune response against the host’s melanoma cells requires immune recognition of target peptides in the context of major histocompatibil- ity complex (MHC) molecules (29–31). This is particularly true for T-cell mediated responses. In addition, co-stimulatory signals are necessary for cell-mediated immunity (32,33). The absence of proper MHC or co-stimulatory molecule expression can prevent an effective antitumor immune response (34). Thus, a melanoma cell that expresses Chapter 35 / Immunogenic Melanoma-Associated Antigens 621 immunogenic MAA may not induce a T-cell mediated antimelanoma response without proper co-stimulation, even if the MAA are recognized by the T-cells. Furthermore, many responses defined as antitumor effector mechanisms can become ineffective or protumorigenic under certain conditions. For example, immune selection pressure could result in outgrowth of resistant tumor variants.

RECOGNITION OF MAA EXPRESSED BY HUMAN MELANOMA CELLS MAA have been detected by in vitro and in vivo cellular reactions and by in vitro serological tests. T- and B-cells that recognize and reject autologous cells have been widely used to identify MAA (35). In serological approaches, both sera from cancer patients and antisera produced in xenogeneic hosts were used as the source of antibody. Antigens Defined by T-Cells Genetic (36), biochemical (37), and immunological (38) approaches have been used to identify MAA recognized by cytolytic T-lymphocytes. MAGE-1 (39) was the first tumor-associated antigen to be identified by a genetic approach. This antigen belongs to a family of at least 12 closely related genes (40) expressed in variable proportions by melanoma cells (41). Expression levels of MAGE antigens, as assessed by poly- merase chain reaction technology, vary considerably among tissue samples and cell lines. MAGE-2 and MAGE-3 are the most common. MAGE-1 has been detected by specific monoclonal antibodies (MAbs) in clinical tumor specimens. The pattern of reactivity observed was heterogeneous within individual specimens (42). When MAGE-1 is expressed, MAGE-2 or MAGE-3 is also expressed, making it feasible to use MAGE-1 and MAGE-3 for therapeutic purposes and reduce the risk of tumor escape caused by emergence of antigen-loss variants (41). Benign and dysplastic nevi, as well as in situ , do not express MAGE genes (43–45). MAGE-1, -2, -3 and -4 are more often expressed by metastatic melanoma than by primary tumors. This difference illustrates the selective activation of certain genes during malignant transformation and progres- sion. Immune responses to the products of these genes may serve as markers of disease status as well as components of a therapeutic antimelanoma response. Melanoblasts, which are precursors of melanocytes, are presumed to have a pheno- typic pattern similar to that of early melanoma cells. MAA such as Melan-A/melanoma antigen recognized by T-cells (MART)-1 (46–48), tyrosinase (49,50), Pme117/gp100 (37,51), gp75/tyrosine-related (TRP)-1 (52), and AIM-2 (53) are expressed by normally differentiated melanocytes and by melanoma cells. These antigens are mela- noma-specific; they are not expressed by other tumor cells. Houghton et al. (54) reported that melanoma cells in early stages of differentiation do not express antigens that are present during the later stages (such as tyrosinase). Using a real-time polymerase chain reaction method, Johansson et al. (55) found that tyrosinase, TRP-1, TRP-2, and MART-1/ Melan-A expression varied from undetectable to highly detectable levels in the different cell lines at different time intervals. Peptides derived from these MAA have been used as targets for immunotherapy in several clinical trials. Although a significant proportion of patients with disease limited to the dermis, subcutaneous tissue, and/or lymphatics developed an immune response, only a minority of patients with metastatic disease had regression of the tumor. Studies by Slingluff et al. (56) suggest that, if a tumor does not express melanocytic differentia- 622 From Melanocytes to Melanoma tion , immunotherapy should be directed against unique tumor antigens, as well as more frequently encountered melanocytic antigens. Antigens Defined by B-Cell Factors Some MAA have been localized on the cell surface, whereas others are found in the cell cytoplasm (57,58). The presence of antinucleolar antibodies in the sera of melanoma patients has also been reported (59). Before the advent of MAbs, numerous studies used patient sera to demonstrate the existence of tumor antigens in melanoma. Use of sero- logical techniques, such as antibody-mediated cytotoxicity, immune adherence, immu- nofluorescence, mixed hemadsorption, complement fixation, radioimmunoassay, and enzyme immunoassay demonstrated the immunological reactivity between these anti- gens and sera from melanoma patients. Based on the humoral crossreactivity and absorp- tion studies, MAA can be grouped into four categories: fetal antigens, common MAA, group-specific antigens with variable expression, and individually specific antigens. Membrane-rich fractions prepared from melanoma tumors showed wide cross- reactivity against sera from patients with malignancies of various histological types. However, absorption of the sera with fetal tissue homogenates revealed that the crossreactivity was caused by fetal antigens. Similar wide crossreactivity was observed with the partially purified spent culture medium of a melanoma cell line, suggesting the expression of these antigens by fetal cells and melanoma cells. Within the last decade, serological expression cloning has been used to identify tumor antigens that elicit a strong antibody response in cancer patients (60). The genes SSX2 (61), NY-ESO-1 (62), and SYCP-1 (63) were identified during serological expression cloning analyses of human cancer. These genes, as well as others such as MAGE, BAGE, and GAGE, have been named “cancer/testis (CT) antigens” (60) because they are ex- pressed predominantly in normal testis and in cancer cells. CT genes are considered ideal target antigens for a cancer vaccine because of their immunogenicity and lack of expression in normal tissues. Of the various CT antigens, NY-ESO-1 has been the most extensively studied as a potential component of cancer vaccines (64). There is a clear distinction between CT genes identified by messenger RNA expres- sion analysis and those identified through immunological methods. The former genes have immunogenic potential, whereas the latter are immunogenic in cancer patients (63). Forty-four CT antigen families have been reported. With the exception of a few genes, such as those of the MAGE family, there is no general evolutionary linkage between CT genes. In addition, protein products of only 19 CT gene families have been demonstrated to elicit an immune response in cancer. The responses were strickly humoral in 13 cases, strictly cellular in 3 cases, and cellular plus humoral in 3 cases (63). Some of the antigenic diversity of human malignant melanoma cells could reflect differences in serological assay techniques, antibody sources (patient sera), and target antigen purity. Conditions for in vitro culture are not the same as those for in vivo tumor growth; the selective pressure of culture conditions may change antigenic expression on or in the cultured melanoma cells. In our experience, the expression level of human MAA fluctuates markedly with passage from one generation to another. Incorporation of exogenous components from growth medium into the membrane of cultured cells is well docu- mented (65), and may influence the results of serological assays. This artifact has been minimized by preparing the target antigen from cells cultured in fetal calf serum-free medium or other medium that is free of potentially immunogenic supplements (66). Chapter 35 / Immunogenic Melanoma-Associated Antigens 623

Hybridoma technology has allowed development of MAbs that recognize tumor- associated antigens expressed in variable levels on autologous cells, melanoma cells, and tumor cells of other histological types. Consistent identification of these antigens— despite varied serological techniques and nonstandardized reagents—confirms exist- ence of the four groups of tumor-associated antigens on melanoma cells. These immunogenic antigens are recognized by allogeneic antibodies and their expression significantly influences the development and progression of melanoma (67). Gangliosides as Melanoma Antigens Gangliosides are carbohydrates that have been used to distinguish different stages of melanocyte/melanoma differentiation (68). These acidic glycolipids contain a hydro- phobic ceramide moiety and are expressed by both normal melanocytes and melanoma cells. GM3, GD3, GM2, GD2, and O-acetyl GD3 are found on the cell membrane of melanocytic cells. Expression of some gangliosides is significantly (>1000-fold, in the case of GD3) upregulated after malignant transformation of melanocytes (69). Regula- tion of ganglioside expression could trigger neoplastic transformation of melanocytes and progression of transformed cells. The diversity of ganglioside expression in neoplastically transformed cells is governed by individual genotypic differences. Changes in ganglioside profile of neoplastically transformed melanocytes correlate with changes in the proliferation, migration, and metastasis of malignant cells (70). Although this chapter is limited to the immunogenic antigens of melanoma, both immunogenic and nonimmunogenic gangliosides play a role in transformation of mel- anocytes and progression of the transformed cells. The presence of autoantibodies to gangliosides has been correlated with an improved prognosis in melanoma patients (71–73). Sialyl Lewis(a) is not immunogenic in melanoma, but its expression has been correlated with melanoma progression (74). Antigens Defined by Antibody Phage Display Library Method A phage-Fab library derived from the B-cells of a melanoma patient in remission after immunotherapy has been used to identify and isolate a 23-kDa glycoprotein (75). The p23 antigen is on the surface and in the cytoplasm of melanoma cells, and it is expressed at high levels by cultured melanoma cell lines, vertical growth-phase primary mela- noma, and metastatic melanoma. The antigen is not expressed in radial growth-phase primary melanoma, nevi, and normal skin. By identifying potentially stage-specific MAA and determining the mechanisms involved in neoplastic progression of trans- formed melanocytes, the antibody phage display library method (76,77) may have thera- peutic and diagnostic potential (78).

ROLE OF IMMUNOGENIC MAA IN MELANOMA PROGRESSION OR INHIBITION Reactions observed in vitro between sera and autologous melanoma cells might also occur in vivo. These reactions, which involve both tumor and host functions, could promote or prevent tumor growth and progression (79). Efforts are underway to increase the quality and magnitude of MAA-specific immune responses to a clinically effective level (80). However, the possibility of antigen loss from tumor cells resulting from immunoselection could result in immune escape and subsequent tumor progression (81–84). 624 From Melanocytes to Melanoma

Several regulatory pathways and molecules may be involved in melanoma antigen silencing, tumor cell differentiation, and the outcome of antitumor immune responses (85–87). Certain melanoma antigens may be modulated by cytokines. In addition, cell and matrix adhesion molecules, growth factors, proteases, gangliosides, and MHC class I and II molecules contribute to the course of malignant melanoma. Antigens expressed on melanoma cells but not on mature melanocytes may be used as markers for the degree of cell differentiation; dedifferentiation of melanoma cells decreases their antigenic similarity to normal melanocytes (88). Different genes responsible for expression of melanoma antigens have been identified in various stages of the disease and may be used as tumor markers for recurrence or progression. Modulation of melanoma antigens has been suggested as a mechanism for tumor progression in the presence of cytolytic T-lymphocytes. This might explain the common finding of Melan-A/MART-1-specific tumor-infiltrating lymphocytes in clinically pro- gressing melanomas, and it might represent a possible pathway for therapeutic interven- tion (89). The antigen-derived T-cell response evoked by early melanoma becomes attenuated with disease progression. A variety of cell adhesion molecules on the surface of melanoma cells may regulate cellular cytotoxicity. The use of MAA, antiantigen autoantibody levels, and immune complexes (IC) has shown promise for detecting disease and for planning and monitoring treatment. Many reports indicate that appearance of antibodies to MAA is associated with improved prognosis. These circulating antimelanoma antibodies can bind to their corresponding antigens and form IC. A clinically interesting example is the hypopigmentation associ- ated with binding of antimelanoma antibodies to normal melanocytes. Serum from pa- tients with vitiligo contains a high titer of naturally occurring antimelanoma antibodies and reportedly induced regression of melanoma metastases in mice. Although the asso- ciation with prognosis is not clear, the appearance of hypopigmentation in patients with melanoma serves as evidence for the activity of antimelanoma antibodies (90). Prognostic Role of Anti-TA90 Immunity in Melanoma Initial studies used allogeneic antibodies to identify a high molecular weight complex in the urine of patients with metastatic melanoma (91,92). This complex comprises multiple subunits, including an immunogenic 90-kDa glycoprotein called TA90 (93). TA90 is a heat-stable antigen with an isoelectric point of 6.1. It is expressed by about 75% of human solid tumors. Immunochemical analysis, including antibody-blocking studies, revealed that TA90 is distinct from known tumor markers such as carcino- embryonic antigen, prostate-specific antigen, CA15-3, D-fetoprotein, and other oncofetal antigens (94). Most cancer patients whose tumors express TA90 have endogenous anti-TA90 anti- bodies of immunoglobulin (Ig)-M and IgG isotypes. In patients with early- or interme- diate-stage melanoma, the immune response to TA90 has been correlated with occult nodal disease and survival (95). In addition, evidence suggests that the endogenous immune response to TA90 might determine the postoperative outcome of patients undergoing surgical therapy for metastatic melanoma (96). Despite the fact that tumor cells have been detected in the blood of patients with metastatic melanoma, many of these patients have enjoyed prolonged survival or cure after surgical resection. This suggests that enhancement of a specific antitumor immune response might prolong survival. In fact, melanoma patients receiving a polyvalent specific active immunothera- Chapter 35 / Immunogenic Melanoma-Associated Antigens 625 peutic (Canvaxin, CancerVax Corp, Carlsbad, CA) that contains TA90 exhibit increased humoral (IgM and IgG) and delayed-type hypersensitivity responses (97,98). In a series of investigations in which patients received Canvaxin after surgical resection of mela- noma, elevated humoral and delayed-type hypersensitivity responses to Canvaxin were associated with prolonged survival (99–101). These observations were the basis for an immunological model to predict the survival of patients who received Canvaxin for regional metastatic melanoma (98). In general, the incidence and level of antibodies to TA90 or other MAA are higher when melanoma is localized than when it is disseminated (102). Surgery and immuno- therapy with autologous or allogeneic vaccines will affect levels of circulating antibody in melanoma patients. Prognostic Role of TA90-Specific Immune Complex in Melanoma When the original high molecular weight glycoprotein complex was purified and used to develop a murine MAb (AD1-40F4) of IgM isotype (103), Western blot analysis revealed that AD1-40F4 recognized a 90-kDa band in urine and serum of cancer patients (93). This epitope is different from the epitope recognized by the autoantibody in the serum of melanoma patients (104). AD1-40F4 was used to develop a TA90-IC assay that discriminated between normal and melanoma sera (105). This assay has a sensitivity of 83% in detecting nodal metastases in melanoma patients undergoing lymphadenec- tomy (94). To test the hypothesis that the postoperative serum level of TA90-IC could have a significant correlation with recurrence and survival in patients with thick primary mela- nomas, postoperative TA90-IC status was correlated with disease-free survival (DFS) and overall survival (OS). Standard prognostic factors for melanoma were then com- pared with TA90-IC. The sensitivity and specificity of TA90-IC for predicting recur- rence were 70% and 85%, respectively. Five-year DFS and OS rates were significantly higher when TA90-IC was negative. At a median follow-up of 25 mo, multivariate analysis identified postoperative TA90-IC status as the only independent predictor of DFS (106). Therefore, a positive postoperative TA90-IC level suggests the presence of micrometastases that may become clinically significant. Because standard prognostic factors, including precise staging of the regional lymph nodes, cannot accurately determine which early-stage melanomas will metastasize, and because TA90-IC status in thick primary melanoma showed a correlation with survival, TA90-IC was also investigated as a prognostic marker for patients with thin primary melanoma. Patients with 1.01- to 2.00-mm primary melanomas and tumor-negative regional lymph nodes were divided into two groups (95). Group 1 comprised 50 patients who died of metastases within 7 yr after complete surgical treatment; group 2 comprised 50 patients who were matched with group 1 by six standard prognostic factors (including tumor thickness, Clark’s level, and presence of ulceration), but who lived at least 10 yr without recurrence. Excluded from study were patients whose stage I melanoma had spread to the regional nodes (stage III disease), either at the time of lymphadenectomy or at any time before the development of distant metastases. This eliminated the possi- bility of studying patients with occult lymph node metastases or locoregional relapse. In addition, this study excluded any patient who received any form of postoperative adju- vant therapy; this eliminated the possible influence of adjuvant therapy on clinical out- comes. All patients in both groups underwent staging of the regional lymph nodes by 626 From Melanocytes to Melanoma complete lymph node dissection or, more recently, by lymphatic mapping and selective dissection of the sentinel lymph node. Postoperative sera from these patients were ana- lyzed for TA90-IC. Standard microscopic examination determined the status of all excised lymph nodes. The incidence of TA90-IC positivity was 82% in group 1 and 18% in group 2 (p < 0.001). Thus, positive TA90-IC level correlated with distant metastasis when melanoma was low- or intermediate risk by standard prognostic factors. TA90-IC has been identified in the sera of numerous melanoma patients. The correlation between TA90-IC and overall survival suggests that subclinical metastasis can be detected before surgical treatment of early-stage melanoma. A subsequent study used postoperative sera to determine TA90-IC levels in patients with melanoma and evaluate their relationship to recurrence and survival (107). Multiple archived serum samples were prospectively collected during postoperative surveillance of 166 patients with American Joint Committee on Cancer stage I, II, or III melanoma. TA90-IC results were correlated with disease recurrence and survival data. The TA90-IC status in the early postoperative period was highly predictive of survival. Five-year OS was 64% for TA90-IC-negative patients and 36% for TA90-IC-positive patients (p = 0.0001). Median OS was 40 mo for TA90-IC-positive groups and 160 mo for the negative groups. TA90-IC-positive patients had a 5-yr DFS of only 24%, compared with 74% for TA90-IC-negative patients (p = 0.0001). Median DFS was 18 mo and 160 mo in TA90-IC-positive and TA90-IC-negative groups, respectively. In a study of 125 stage IV melanoma patients undergoing complete resection of distant metastases, postopera- tive TA90-IC positivity was again the most important prognostic variable for DFS (108). Thus, there is ample evidence to suggest that a positive TA90-IC assay strongly corre- lates with recurrence after surgical resection of American Joint Committee on Cancer stage II, III, and IV melanoma.

CONCLUSIONS AND PERSPECTIVES Tumor immunology represents a complex interplay of many different factors that affect tumor cell growth. Results of various investigations of immune responses against melanoma antigens indicate that both cellular and humoral immune responses to MAA are critical to the development and control of progressive disease (109). Clinical reports of spontaneous regression of melanoma and the improved outcomes seen in patients receiving experimental vaccines are promising indications of the significant impact that immunotherapy may have on the future treatment of melanoma. However, development of recurrence and the progression of treated disease suggest that melanoma cells may escape immune destruction by losing or downregulating expression of MAA targets. Continued studies of MAA-based interactions may allow the development of antigen- targeted immunotherapy that prevents the malignant transformation of melanocytes to melanoma.

REFERENCES 1. Barnetson RS, Halliday GM. Regression in skin tumours: a common phenomenon. Australas J Dermatol 1997;38:S63–S65. 2. Printz C. Spontaneous regression of melanoma may offer insight into cancer immunology. J Natl Cancer Inst 2001;93:1047–1048. 3. Cochran AJ, Diehl V, Stjernsward J. Regression of primary malignant melanoma associated with a good prognosis despite metastasis to lymph nodes. Rev Eur Etud Clin Biol 1970;15:969–972. Chapter 35 / Immunogenic Melanoma-Associated Antigens 627

4. Morton DL, Eilber FR, Malmgren RA, et al. Immunological factors which influence response to immunotherapy in malignant melanoma. Surgery 1970;68:158–164. 5. Morton DL, Malmgren RA, Holmes EC, et al. Demonstration of antibodies against human malignant melanoma by immunofluorescence. Surgery 1968;64:233–240. 6. Summer WC, Foraker AG. Spontaneous regression of human melanoma: clinical and experimental studies. Cancer 1960;13:79–81. 7. Movassagh M, Spatz A, Davoust J, et al. Selective accumulation of mature DC-Lamp+ dendritic cells in tumor sites is associated with efficient T-cell–mediated antitumor response and control of meta- static dissemination in melanoma. Cancer Res 2004;64:2192–2198. 8. Mihm MC Jr, Clemente CG, Cascinelli N. Tumor infiltrating lymphocytes in lymph node melanoma metastases: a histopathologic prognostic indicator and an expression of local immune response. Lab Invest 1996;74:43–47. 9. Hersey P, Murray E, Grace J, McCarthy WH. Current research on immunopathology of melanoma: analysis of lymphocyte populations in relation to antigen expression and histological features of melanoma. Pathology 1985;17:385–391. 10. Steiner A, Wolf C, Pehamberger H, Wolff K. Late metastases of cutaneous malignant melanoma. Br J Dermatol 1986;114:737–740. 11. Koh HK, Sober AJ, Fitzpatrick TB. Late recurrence (beyond ten years) of cutaneous malignant melanoma. Report of two cases and a review of the literature. JAMA 1984;251:1859–1862. 12. Lejeune FJ. The impact of surgery on the course of melanoma. Recent Results Cancer Res 2002;160:151–157. 13. Shields JD, Borsetti M, Rigby H, et al. Lymphatic density and metastatic spread in human malignant melanoma. Br J Cancer 2004;90:693–700. 14. Nathanson SD. Insights into the mechanisms of lymph node metastasis. Cancer 2003;98:413–423. 15. Fallowfield ME, Cook MG. Lymphatics in primary cutaneous melanoma. Am J Surg Pathol 1990;14:370–374. 16. Griffiths JD. Circulating cancer cells in surgery. Lond Clin Med J 1966;7:65–71. 17. Roberts SS, Cole WH. Cancer cells in the circulating blood. Semin Int 1961;10:2–10. 18. Keilholz U, Goldin-Lang P, Bechrakis NE, et al. Quantitative detection of circulating tumor cells in cutaneous and ocular melanoma and quality assessment by real-time reverse transcriptase-poly- merase chain reaction. Clin Cancer Res 2004;10:1605–1612. 19. Mocellin S, Del Fiore P, Guarnieri L, et al. Molecular detection of circulating tumor cells is an independent prognostic factor in patients with high-risk cutaneous melanoma. Int J Cancer 2004;111:741–745. 20. Palmieri G, Ascierto PA, Perrone F, et al. Prognostic value of circulating melanoma cells detected by reverse transcriptase-polymerase chain reaction. J Clin Oncol 2003;21:767–773. 21. Tsubura E, Yamashita T, Sone S. Inhibition of the arrest of hematogenously disseminated tumor cells. Cancer Metastasis Rev 1983;2:223–237. 22. Morton DL, Sparks FC, Haskell CM. Oncology. In: Swartz SI, Shire CT, Spencer FC, Storer EH, eds. Principles of Surgery, 3rd ed. McGraw Hill, New York, NY: 1979. 23. Vile RG, Chong H, Dorudi S. The immunosurveillance of cancer. In: Tumor Immunology: Immuno- therapy and Cancer Vaccines. Dalgleish AG, Browning M, eds. Cambridge University Press, Cam- bridge, UK: 1996. 24. Wang RF. Human tumor antigens: implications for cancer vaccine development. J Mol Med 1999;77:640–655. 25. Faries MB, Morton DL. Melanoma: is immunotherapy of benefit? Adv Surg 2003;37:139–169. 26. Morton DL, Ollila DW, Hsueh EC, Essner R, Gupta RK. Cytoreductive surgery and adjuvant immunotherapy: a new management paradigm for metastatic melanoma. CA Cancer J Clin 1999;49:101–116. 27. Bitton RJ. Cancer vaccines: a critical review on clinical impact. Curr Opin Mol Ther 2004;6:17–26. 28. Pawelec G. Tumor escape: anti-tumor effectors too much of a good thing? Cancer Immunol Immunother 2004;53:262–274. 29. Parkhurst MR, Riley JP, Robbins PF, Rosenberg SA. Induction of CD4+ Th1 lymphocytes that recognize known and novel class II MHC restricted epitopes from the melanoma antigen gp100 by stimulation with recombinant protein. J Immunother 2004;27:79–91. 30. Mandic M, Almunia C, Vicel S, et al. The alternative open reading frame of LAGE-1 gives rise to multiple promiscuous HLA-DR-restricted epitopes recognized by T-helper 1-type tumor-reactive CD4+ T cells. Cancer Res 2003;63:6506–6515. 628 From Melanocytes to Melanoma

31. Maeurer MJ, Storkus WJ, Kirkwood JM, Lotze MT. New treatment options for patients with mela- noma: review of melanoma-derived T-cell epitope-based peptide vaccines. Melanoma Res 1996;6:11–24. 32. Schwartz RH. Acquisition of immunologic self-tolerance. Cell 1989;57:1073–1081. 33. Janeway CA Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol 1989;54 Pt 1:1–13. 34. Chen L, Ashe S, Brady WA, et al. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 1992;71:1093–1102. 35. Ramirez-Montagut T, Turk MJ, Wolchok JD, Guevara-Patino JA, Houghton AN. Immunity to mela- noma: unraveling the relation of tumor immunity and autoimmunity. Oncogene 2003;22:3180–3187. 36. Coulie PG, Lehmann F, Lethe B, et al. A mutated intron sequence codes for an antigenic peptide recognized by cytolytic T lymphocytes on a human melanoma. Proc Natl Acad Sci USA 1995;92:7976–7980. 37. Cox AL, Skipper J, Chen Y, et al. Identification of a peptide recognized by five melanoma-specific human cytotoxic lines. Science 1994;264:716–719. 38. Mukherji B, MacAlister TJ. Clonal analysis of cytotoxic T cell response against human melanoma. J Exp Med 1983;158:240–245. 39. van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991;254:1643–1647. 40. De Plaen E, Arden K, Traversari C, et al. Structure, chromosomal localization, and expression of 12 genes of the MAGE family. Immunogenetics 1994;40:360–369. 41. Coulie PG. Human tumor antigens recognized by cytolytic T lymphocytes. In: Tumor Immunology: Immunotherapy and Cancer Vaccines. Cambridge University Press, New York, NY: 1996, pp. 95–125. 42. Zuber M, Spagnoli GC, Kocher T, et al. Heterogeneity of melanoma antigen-1 (MAGE-1) gene and protein expression in malignant melanoma. Eur Surg Res 1997;29:403–410. 43. Van den Eynde B, Peeters O, De Backer O, Gaugler B, Lucas S, Boon T. A new family of genes coding for an antigen recognized by autologous cytolytic T lymphocytes on a human melanoma. J Exp Med 1995;182:689–698. 44. Brasseur F, Rimoldi D, Lienard D, et al. Expression of MAGE genes in primary and metastatic cutaneous melanoma. Int J Cancer 1995;63:375–80. 45. Dalerba P, Ricci A, Russo V, et al. High homogeneity of MAGE, BAGE, GAGE, tyrosinase and Melan-A/MART-1 in clusters of multiple simultaneous metastases of human mela- noma: implications for protocol design of therapeutic antigen-specific vaccination strategies. Int J Cancer 1998;77:200–204. 46. Kawakami Y, Rosenberg SA. Human tumor antigens recognized by T-cells. Immunol Res 1997;16:313–339. 47. Murer K, Urosevic M, Willers J, et al. Expression of Melan-A/MART-1 in primary melanoma cell cultures has prognostic implication in metastatic melanoma patients. Melanoma Res 2004; 14:257–262. 48. Lee PP, Yee C, Savage PA, et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat Med 1999;5:677–685. 49. Lotz C, Ferreira EA, Drexler I, et al. Partial tyrosinase-specific self tolerance by HLA-A*0201- restricted cytotoxic T lymphocytes in mice and man. Int J Cancer 2004;108:571–579. 50. Wang RF, Appella E, Kawakami Y, Kang X, Rosenberg SA. Identification of TRP-2 as a human tumor antigen recognized by cytotoxic T lymphocytes. J Exp Med 1996;184:2207–2216. 51. Bakker AB, Schreurs MW, de Boer AJ, et al. Melanocyte lineage-specific antigen gp100 is recog- nized by melanoma-derived tumor-infiltrating lymphocytes. J Exp Med 1994;179:1005–1009. 52. Vijayasaradhi S, Bouchard B, Houghton AN. The melanoma antigen gp75 is the human homologue of the mouse b (brown) gene product. Exp Med 1990;171:1375–1380. 53. Harada M, Li YF, El-Gamil M, Rosenberg SA, Robbins PF. Use of an in vitro immunoselected tumor line to identify shared melanoma antigens recognized by HLA-A*0201-restricted T cells. Cancer Res 2001;61:1089–1094. 54. Houghton AN, Vijayasardhi S, Bouchard B, Nazftzger C, Hara I, Chapman PB. Recognition of autoantigens by patients with melanoma. Ann NY Acad Sci 1993;690:59–68. 55. Johansson M, Takasaki A, Lenner L, Arstrand K, Kagedal B. Quantitative relationships between pigment-related mRNA and biochemical melanoma markers in melanoma cell lines. Melanoma Res 2002;12:193–200. Chapter 35 / Immunogenic Melanoma-Associated Antigens 629

56. Slingluff CL Jr, Colella TA, Thompson L, et al. Melanomas with concordant loss of multiple melanocytic differentiation proteins: immune escape that may be overcome by targeting unique or undefined antigens. Cancer Immunol Immunother 2000;48:661–672. 57. Wolchok JD, Weber JS, Houghton AN, Livingston PO. Melanoma vaccines. In: Balch CM, Houghton AN, Sober AJ, Soong SJ, eds. Cutaneous Melanoma, 4th ed. Quality Medical Publishing, St Louis, MO: 2003. 58. Bar-Dayan Y, Barshack I, Blank M, et al. Antibodies to the cytoplasm, cell membrane and nuclear membrane of malignant neoplasms in pooled normal human polyspecific immunoglobulin G. Int J Oncol 1999;15:1091–1096. 59. Thomas PJ, Kaur JS, Aitcheson CT, Robinson WA, Tan EM. Antinuclear, antinucleolar, and anticytoplasmic antibodies in patients with malignant melanoma. Cancer Res 1983;43:1372–1380. 60. Sahin U, Tureci O, Schmitt H, et al. Human neoplasms elicit multiple specific immune responses in the autologous host. Proc Natl Acad Sci USA 1995;92:11,810–11,813. 61. Chen YT, Scanlan MJ, Sahin U, et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci USA 1997;94:1914–1918. 62. Tureci O, Sahin U, Zwick C, Koslowski M, Seitz G, Pfreundschuh M. Identification of a meiosis- specific protein as a member of the class of cancer/testis antigens. Proc Natl Acad Sci USA 1998;95:5211–5216. 63. Scanlon MJ, Simpson AJG, Old LJ. The cancer/testis genes: review, standardization, and commen- tary. Cancer Immunity 2004;4:1–15. 64. Old LJ. Cancer Vaccine Collaborative 2002: opening address. Cancer Immun 2003;3:1. 65. Natali PG, Wilson BS, Imai K, Bigotti A, Ferrone S. Tissue distribution, molecular profile, and shedding of a cytoplasmic antigen identified by the monoclonal antibody 465.12S to human mela- noma cells. Cancer Res 1982;42:583–589. 66. Euhus DM, Gupta RK, Morton DL. Induction of antibodies to a tumor-associated antigen by immu- nization with a whole melanoma cell vaccine. Cancer Immunol Immunother 1989;29:247–254. 67. Gupta RK, Morton DL. Clinical significance of tumor-associated antigens and antitumor antibodies in human malignant melanoma. In: Reisfeld RA, Ferrone S, eds. Melanoma Antigens and Antibodies. Plenum, New York, NY: 1982, pp. 139–172. 68. Houghton AN, Wolchok JD. Immunology. In: Balch CH, Sober AJ, Soon SJ, eds. Cutaneous Mela- noma. Quality Medical Publishing, St. Louis, MO. 2003 pp. 659–668. 69. Albino AP, Sozzi G, Nanus DM, Jhanwar SC, Houghton AN. Malignant transformation of human melanocytes: induction of a complete melanoma phenotype and genotype. Oncogene 1992;7: 2315–2321. 70. Ravindranath MH, Tuchida T, Irie RF. Diversity of ganglioside expression in human melanoma: In: Oettgen HF, ed. Gangliosides and Cancer. VCH Publishers, New York, NY: 1989, pp 80–91. 71. Ravindranath MH, Gonzales AM, Nishimoto K, Tam WY, Soh D, Morton DL. Immunology of gangliosides. Indian J Exp Biol 2000;38:301–312. 72. Livingston PO, Wong GY, Adluri S, et al. Improved survival in stage III melanoma patients with GM2 antibodies: a randomized trial of adjuvant vaccination with GM2 ganglioside. J Clin Oncol 1994;12:1036–1044. 73. Ravindranath MH, Morton DL. Immunogenicity of membrane-bound gangliosides in viable whole- cell vaccines. Cancer Invest 1997;15:491–499. 74. Kageshita T, Hirai S, Kimura T, Hanai N, Ohta S, Ono T. Association between sialyl Lewis(a) expression and tumor progression in melanoma. Cancer Res 1995;55:1748–1751. 75. Li J, Pereira S, Van Belle P, et al. Isolation of the melanoma-associated antigen p23 using antibody phage display. J Immunol 2001;166:432–438. 76. Cai X, Garen A. Anti-melanoma antibodies from melanoma patients immunized with genetically modified autologous tumor cells: selection of specific antibodies from single-chain Fv fusion phage libraries. Proc Natl Acad Sci USA 1995;92:6537–6541. 77. Kupsch JM, Tidman NH, Kang NV, et al. Isolation of human tumor-specific antibodies by selection of an antibody phage library on melanoma cells. Clin Cancer Res 1999;5:925–931. 78. Desai SA, Wang X, Noronha EJ, Kageshita T, Ferrone S. Characterization of human anti-high molecular weight-melanoma-associated antigen single-chain Fv fragments isolated from a phage display antibody library. Cancer Res 1998;58:2417–2425. 79. Hanahan D, Lanzavecchia A, Mihich E. Fourteenth Annual Pezcoller Symposium: the novel dichotomy of immune interactions with tumors. Cancer Res 2003;63:3005–3008. 630 From Melanocytes to Melanoma

80. Belardelli F, Ferrantini M, Parmiani G, Schlom J, Garaci E. International meeting on cancer vaccines: how can we enhance efficacy of therapeutic vaccines? Cancer Res 2004;64:6827–6830. 81. Cormier JN, Hijazi YM, Abati A, et al. Heterogeneous expression of melanoma-associated antigens and HLA-A2 in metastatic melanoma in vivo. Int J Cancer 1998;75:517–524. 82. Khong HT, Wang QJ, Rosenberg SA. Identification of multiple antigens recognized by tumor- infiltrating lymphocytes from a single patient: tumor escape by antigen loss and loss of MHC expres- sion. J Immunother 2004;27:184–190. 83. Radford KJ, Mallesch J, Hersey P. Suppression of human melanoma cell growth and metastasis by the melanoma-associated antigen CD63 (ME491). Int J Cancer 1995;62:631–635. 84. Kondoh M, Ueda M, Ichihashi M, Mishima Y. Decreased expression of human melanoma-associated antigen ME491 along the progression of melanoma pre-canceroses to invasive and metastatic mela- nomas. Melanoma Res 1993;3:241–245. 85. Hoek K, Rimm DL, Williams KR, et al. Expression profiling reveals novel pathways in the transfor- mation of melanocytes to melanomas. Cancer Res 2004;64:5270–5282. 86. Du J, Miller AJ, Widlund HR, Horstmann MA, Ramaswamy S, Fisher DE. MLANA/MART1 and SILV/PMEL17/GP100 are transcriptionally regulated by MITF in melanocytes and melanoma. Am J Pathol 2003;163:333–343. 87. Durda PJ, Dunn IS, Rose LB, et al. Induction of “antigen silencing” in melanomas by oncostatin M: down-modulation of melanocyte antigen expression. Mol Cancer Res 2003;1:411–419. 88. Merimsky O, Shoenfeld Y, Chaitchik S, Yecheskel G, Fishman P. Antigens and antibodies in malig- nant melanoma. Tumour Biol 1994;15:188–202. 89. Ramirez-Montagut T, Andrews DM, Ihara A, et al. Melanoma antigen recognition by tumour-infil- trating T lymphocytes (TIL): effect of differential expression of melan-A/MART-1. Clin Exp Immunol 2000;119:11–18. 90. Merimsky O, Shoenfeld Y, Yecheskel G, Chaitchik S, Azizi E, Fishman P. Vitiligo- and melanoma- associated hypopigmentation: a similar appearance but a different mechanism. Cancer Immunol Immunother 1994;38:411–416. 91. Rote NS, Gupta RK, Morton DL. Tumor-associated antigens detected by autologous sera in urine of patients with solid neoplasms. J Surg Res 1980;29:18–22. 92. Rote NS, Gupta RK, Morton DL. Determination of incidence and partial characterization of tumor- associated antigens found in the urine of patients bearing solid tumors. Int J Cancer 1980;26:203–210. 93. Euhus DM, Gupta RK, Morton DL. Characterization of a 90-100 kDa tumor-associated antigen in the sera of melanoma patients. Int J Cancer 1990;45:1065–1070. 94. Kelley MC, Jones RC, Gupta RK, et al. Tumor-associated antigen TA-90 immune complex assay predicts subclinical metastasis and survival for patients with early stage melanoma. Cancer 1998;83:1355–1361. 95. Litvak DA, Gupta RK, Yee R, Wanek LA, Ye W, Morton DL. Endogenous immune response to early- and intermediate-stage melanoma is correlated with outcomes and is independent of locoregional relapse and standard prognostic factors. J Am Coll Surg 2004;198:227–235. 96. Hsueh EC, Gupta RK, Yee R, Leopoldo ZC, Qi K, Morton DL. Does endogenous immune response determine the outcome of surgical therapy for metastatic melanoma? Ann Surg Oncol 2000; 7:232–238. 97. Euhus DM, Gupta RK, Morton DL. Induction of antibodies to a tumor-associated antigen by immu- nization with a whole melanoma cell vaccine. Cancer Immunol Immunother 1989;29:247–254. 98. Jones RC, Kelley MC, Gupta RK, et al. Immune response to polyvalent melanoma cell vaccine in AJCC stage III melanoma: an immunologic survival model. Ann Surg Oncol 1996;3:437–445. 99. Chung MH, Gupta RK, Hsueh E, et al. Humoral immune response to a therapeutic polyvalent cancer vaccine after complete resection of thick primary melanoma and sentinel lymphadenectomy. J Clin Oncol 2003;21:313–319. 100. DiFronzo LA, Gupta RK, Essner R, et al. Enhanced humoral immune response correlates with improved disease-free and overall survival in American Joint Committee on Cancer stage II mela- noma patients receiving adjuvant polyvalent vaccine. J Clin Oncol 2002;20:3242–3248. 101. Hsueh EC, Gupta RK, Qi K, Morton DL. Correlation of specific immune responses with survival in melanoma patients with distant metastases receiving polyvalent melanoma cell vaccine. J Clin Oncol 1998;16:2913–2920. 102. Wong JH, Gupta RK, Morton DL. Serial determinations of melanoma tumor-associated antigen and antibody in patients with stage I melanoma. Arch Surg 1986;121:1342–1345. Chapter 35 / Immunogenic Melanoma-Associated Antigens 631

103. Euhus DM, Gupta RK, Morton DL. Detection of a tumor-associated glycoprotein antigen in serum and urine of melanoma patients by murine monoclonal antibody (AD1-40F4) in enzyme immunoas- say. J Clin Lab Anal 1989;3:184–190. 104. Euhus DM, Gupta RK, Morton DL. Association between allo-immunoreactive and xeno-immunore- active subunits of a glycoprotein tumor-associated antigen. Cancer Immunol Immunother 1990;32:214–220. 105. Gupta RK, Morton DL. Monoclonal antibody-based ELISA to detect glycoprotein tumor-associated- antigen specific immune complexes in cancer patients. J Clin Lab Anal 1992;6:329–336. 106. Chung MH, Gupta RK, Essner R, Ye W, Yee R, Morton DL. Serum TA90 immune complex assay can predict outcome after resection of thick (> or =4 mm) primary melanoma and sentinel lym- phadenectomy. Ann Surg Oncol 2002;9:120–126. 107. Kelley MC, Gupta RK, Hsueh EC, Yee R, Stern S, Morton DL. Tumor-associated antigen TA90 immune complex assay predicts recurrence and survival after surgical treatment of stage I-III mela- noma. J Clin Oncol 2001;19:1176–1182. 108. Hsueh EC, Gupta RK, Qi K, Yee R, Leopoldo ZC, Morton DL. TA90 immune complex predicts survival following surgery and adjuvant vaccine immunotherapy for stage IV melanoma. Cancer J Sci Am 1997;3:364–370. 109. Halliday GM. Skin immunity and melanoma development. In: Thompson JF, Morton DL, Kroon BR, eds. Textbook of Melanoma. Martin Dunitz, Taylor and Francis Group, New York, NY: 2004, pp. 25–42.