Abstract
It recently has become clear that multiple molecular subtypes of melanoma likely exist that may be associated with clinical response to defined therapeutic modalities. Gene expression profiling has revealed a signature that is associated with clinical benefit to melanoma vaccines, with preliminary work suggesting a correlation with response to other immunotherapy agents as well. Activating mutations in B-Raf and c-kit are associated with clinical response to the specific kinase inhibitors PLX4032 and imatinib, respectively. Several other signal transduction pathways have been found to be constitutively active or mutated in other subsets of melanoma tumors that are potentially targetable with new agents. Together, these emerging data suggest the evolution of a new paradigm in melanoma therapy in which molecular analysis of the tumor will be utilized to assign the most appropriate therapeutic modality for each individual patient, to maximize therapeutic success.
Evidence for existence of biologic subsets of melanoma
Only two FDA approved drugs are available to treat patients with metastatic melanoma, the chemotherapeutic agent dacarbazine (approved in 1976) and the immunomodulatory cytokine interleukin-2 (IL-2; approved in 1998). Each of these produces response rates of less than 15% in unselected patients. While it has been argued that melanoma is simply an aggressive tumor generally resistant to therapies, it is of interest that there is not cross-resistance between dacarbazine and IL-2 (1). Some patients who progress after treatment with dacarbazine clearly can respond to IL-2, and vice versa. In addition, combination regimens of chemotherapy and cytokines (biochemotherapy designs) appear to show additive but not synergistic activity, with increased response rates but no improvement in overall survival observed in randomized trials (2). Together, these observations have suggested that there may be subsets of melanoma patients with biologic characteristics that render them susceptible to the therapeutic effect of one modality (chemotherapy) versus another (immunotherapy). However, all histological subtypes of melanoma (superficial spreading, acral lentiginous, nodular, lentigo maligna, and mucosal) have traditionally been clinically managed identically.
Molecular evidence pointing to the existence of clinically meaningful subsets of melanoma came from the comparative genomic hybridization and systematic oncogene mutation analyses studies of Bastian and colleagues (3). In those studies, melanomas arising in a context of sun-damaged skin, non-sun-damaged skin, acral surfaces, or mucosal regions were found to have distinct major molecular aberrations. In particular, mutations in B-Raf were most frequently found in lesions from non-sun-damaged skin. Amplifications in CCND1 and CDK4 (downstream mediators of cell cycle progression in the Ras pathway) were found in lesions that lacked upstream mutations in N-Ras or B-Raf. In addition, amplifications in the c-kit gene locus were found in a significant proportion of acral and mucosal lesions. Thus, rather than classical histological subcategorization of melanoma lesions, these comprehensive results suggested that molecular subtyping of melanoma may have greater practical utility, as drugs that specifically target receptor tyrosine kinases, downstream kinases, and other signaling proteins become available.
Gene expression profiling as a predictive biomarker for response to melanoma vaccines
The best clinical responses of metastatic melanoma to immunotherapeutic agents such as IL-2, IFN-α2b, and experimental cancer vaccines are around 10-15%. In the case of vaccines, it has been possible to investigate potential correlations between clinical response in terms of tumor regression and the magnitude of the specific T cell response induced by the vaccine as measured in the peripheral blood. While such correlations have been observed in some studies, clinical responses have clearly been seen in patients with frequencies of such T cells below the limit of detection using standard assays (4), and conversely, complete lack of clinical benefit has been seen in patients with very high T cell frequencies (5). This apparent paradox led investigators to perform a systematic analysis of the tumor site to probe for factors in the tumor microenvironment that may determine clinical outcome to melanoma vaccines. Using Affymetrix gene expression profiling, clinical benefit was seen in a subset of patients who showed an “inflamed” tumor microenvironment at baseline (6, 7). Metastases of this type show expression of an array of chemokines predicted to be capable of recruiting activated T cells into the tumor site; differential expression of these chemokines has been confirmed at the protein level and/or by qRT-PCR. Using an in vivo xenograft model, preferential recruitment of CD8+ effector T cells into those tumors producing high levels of chemokines was confirmed (8). These results suggest that one major determining factor for clinical response to melanoma vaccines is whether T cell trafficking into the tumor microenvironment can be supported in individual cases, thus allowing T cell access to tumor cells and an opportunity for immune-mediated tumor cell killing. A similar immune signature suggesting an “inflamed” phenotype has been seen in two additional vaccine trials in association with favorable clinical benefit (9, 10).
It may seem on the surface paradoxical that a subset of tumors shows spontaneous inflammation that includes activated CD8+ T cells. In a limited number of patients with sufficient fresh tissue for analysis, peptide/HLA-A2 tetramer staining has confirmed that a subset of these T cells in fact recognizes tumor antigens (11-14). Why these tumors are not rejected spontaneously may be explained by T cell dysfunction in the tumor microenvironment. Ex vivo analysis of CD8+ T cells from melanoma metastases has shown minimal expression of cytotoxic granule proteins and defective cytokine production in response to specific antigen in vitro. The T cell-rich tumors also express the highest level of defined immune inhibitory factors (15). These include the tryptophan-catabolizing enzyme indoleamine-2,3-dioxygenase (IDO), which has been implicated in maternal/fetal tolerance (16); the ligand PD-L1/B7-H1, which engages an inhibitory receptor on activated T cells called PD-1 (17); and the presence of regulatory T cells expressing the CD4+CD25+FoxP3+ phenotype, which have been shown to mediate extrinsic suppression of activated T cells in the tumor setting as well as other immune responses in vivo (18). In addition, these tumors lack meaningful levels of expression of the T cell costimulatory ligands B7-1 or B7-2, arguing that classical T cell anergy may also be operational (19).
Together, these observations are beginning to have several important implications. First, the presence of the “inflamed” tumor phenotype might serve as a viable predictive biomarker for clinical benefit to melanoma vaccines. This notion is currently being explored in prospective multicenter studies of the MAGE-3 protein vaccine developed by GSK-Bio (20). Second, the presence of defined immune inhibitory mechanisms in those inflamed tumors suggests that, as an alternative to vaccine approaches aiming to increase the number of functional T cells that enter the tumor, it may be possible to interfere with the function of immune suppressive pathways. To this end, blockade of IDO with small molecule inhibitors (21, 22), interference with PD-1/PD-L1 interactions with specific monoclonal antibodies (23, 24), depletion of Tregs by targeting CD25 (25), and reversal of T cell anergy through forced homeostatic proliferation (26) have all shown efficacy in preclinical in vivo tumor models. Moreover, recent clinical experience has begun to be generated with several of these approaches. The IDO inhibitor 1-methyltryptophan is currently undergoing phase I clinical testing, and a newer more potent IDO inhibitor has just entered the clinic (27). Impressive phase II results with an anti-PD-1 mAb were presented at the 2010 ASCO annual meeting, in which approximately 30% of patients with advanced melanoma, renal cell carcinoma, and non-small-cell lung cancer showed clinical responses (28). Preliminary biomarker studies in a subset of patients with available tissue for analysis has suggested that clinical benefit may be enriched in the patients with tumors showing high cell surface staining for PD-L1. Depletion of Tregs has been pursued with denileukin diftitox (29), an IL-2-diptheria toxin fusion protein, and with daclizumab, an anti-CD25 mAb (30). Interestingly, clinical responses with denileukin diftitox as a single agent have been reported in melanoma (31). Finally, homeostatic proliferation of T cells, driven by their transfer into lymphopenic hosts, has been found to markedly increase the clinical efficacy of adoptively transferred autologous tumor-infiltrating lymphocytes in melanoma (32). Together, these observations firmly support the continued study of the tumor microenvironment for clues to improve the effector phase of the anti-tumor T cell response toward improved tumor rejection in patients.
In terms of the predictive biomarker implications of these studies, one obvious open question is whether clinical benefit from other immunotherapeutic approaches beyond vaccines might also be associated with the “inflamed” melanoma tumor microenvironment phenotype. In fact, preliminary results presented at the ASCO 2009 annual meeting are consistent with this notion. Atkins and colleagues reported that tumors with expression of a set of chemokines and cytokines were more likely to respond after treatment with IL-2 (33). In addition, Hamid and colleagues reported that clinical response to the anti-CTLA-4 mAb ipilimumab were more likely to occur in patients with tumors expressing several immunoregulatory molecules (34). Together, these observations suggest that an ongoing dialogue between the tumor and the host immune response might be a prerequisite for clinical benefit to several classes of immunotherapeutic interventions, a concept that should be evaluated in prospective clinical studies of those agents.
Kinase mutations and clinical response to kinase inhibitors
B-Raf
The identification of activating mutations in B-Raf as a common genetic alteration in melanoma (35) rapidly led to the hypothesis that kinase inhibitors that target B-Raf might have therapeutic utility. Over 90% of B-Raf mutations in melanoma involve a substitution of glutamate for valine at position 600 (the V600E mutation), and over 50% of melanomas carry such a mutation. A wave of enthusiasm embraced the first agent with potential inhibitory activity against Raf family kinases, sorafenib. Despite the lack of meaningful clinical responses among melanoma patients treated on the phase I/II studies of sorafenib as a single agent (36), an unusually high response rate to carboplatin and paclitaxel combined with sorafenib sustained interest in the drug for melanoma (37). These observations led to two phase III trials of carboplatin and paclitaxel with or without sorafenib in patients with metastatic melanoma, either in the first line or second line setting. Unfortunately, there were no significant differences in clinical outcome in either study (38), which temporarily dampened enthusiasm for the concept of Raf blockade in this disease. The clinical activity of sorafenib in other cancers, such as renal cell carcinoma, is presumed to be mediated through inhibition of other kinases, including the VEGF receptor tyrosine kinase. Other clinical trials aiming to block Ras pathway signaling at other levels, including farnesyltransferase inhibitors (aiming to target Ras proteins directly) and MEK inhibitors (targeting the kinase downstream from Raf), also showed disappointing clinical activity in melanoma (39).
However, other small molecule inhibitors with more potent activity against mutant B-Raf had continued in development. PLX4032 was reported to have 8-fold greater activity against mutant B-Raf over wild-type Raf, showing inhibition in vitro at nanomolar concentrations. A phase I study with an expansion cohort in melanoma was conducted and recently reported (40). Of the 48 V600E B-Raf mutated melanoma patients treated at the recommended phase II dose, 34 partial and 3 complete responses were observed. In contrast, the 5 patients with melanomas expressing wildtype B-Raf had no clinical response. These impressive results have provided the first evidence that an agent targeting a commonly mutated signaling protein in melanoma can exert meaningful clinical activity. A phase III study of PLX4032 is ongoing to determine whether an overall survival benefit can be observed.
c-kit
Along with the report by Bastian and colleagues that the c-kit gene is amplified in a subset of melanomas, activating mutations in c-kit have been observed. Interestingly, these have been seen in around 30% of tumors from mucosal and acral sites, as well as a minority of patients with melanomas arising out of sun damaged skin (41, 42). The spectrum of mutations identified to date parallel those reported in gastrointestinal stromal tumors, which are associated with clinical response to c-kit inhibitors such as imatinib (43). Based on these observations, imatinib has been investigated clinically in melanoma patients bearing c-kit mutations, and studies with nilotinib and other kinase inhibitors have recently been initiated. Several case reports have now been published (44-47), revealing at least 10 clinical responders in total. In contrast, clinical testing of imatinib in unselected melanoma patients revealed minimal clinical activity (48, 49). Thus, it is very likely that a clinically relevant response rate will be observed in the subset of melanoma patients with tumors that have specific activating mutations in c-kit. This notion is currently being examined systematically in a series of prospective phase II and phase III clinical trials.
Molecular features associated with response to chemotherapy
The fact that clinical response to kinase inhibitors might be predicted based on the presence of activating mutations in the relevant kinase has raised the question of whether the activity of traditional chemotherapeutic agents might also be associated with a predictive biomarker. Sensitivity versus resistance to alkylating agents such as dacarbazine or temozolomide might be predicted to be inversely correlated with expression of DNA repair enzymes, such as O6-methylguanine methyltransferase (MGMT). Indeed, methylation and presumed silencing of the MGMT gene has been shown to be associated with a favorable clinical response of glioblastoma to temozolomide plus radiation (50). With this premise as a foundation, Tawbi and colleagues recently reported on a study of molecular profiling in melanoma and association with outcome in 21 patients treated with dacarbazine. Using a combination of gene expression profiling and analysis of gene locus methylation status, they identified a 9 gene predictor (51). Interestingly, some of these genes encode signaling proteins (RasSF4) or immunoregulatory molecules (NKG7). It is noteworthy that MGMT did not emerge as a candidate gene in this study. This is consistent with the lack of added clinical benefit with the addition of the MGMT inhibitor O6-benzylguanine in melanoma (52), and suggests that alternative resistance mechanisms of melanoma to alkylating agents likely are dominant. While still early in development, these initial observations support continued investigation of potential predictive biomarkers for clinical benefit to standard chemotherapeutic agents in this disease.
New pathways showing heterogeneity among individual melanoma patients
In addition to mutations that lead to activation of the Ras pathway, other parallel signaling pathways have been found to be constitutively activated in subsets of melanoma tumors and could lead to new therapeutic approaches in select groups of patients. The Notch pathway has been reported to be activated in many melanomas, apparently via ligand and receptor overexpression rather than through mutation (53). Notch signaling is mediated, in part, through proteolytic cleavage by an enzyme called gamma secretase, which liberates the intracellular domain of Notch to participate in transcriptional regulation. Gamma secretase inhibitors (GSIs) have been developed for clinical exploration as a strategy to inhibit Notch pathway signaling in patients. Results of a phase I study of a GSI were presented at the ASCO 2010 annual meeting, with two melanoma patients showing clinical responses (54). Phase II studies of this agent in melanoma are in the planning stages. Activating mutations in PI3 kinase have been reported in a minor subset of melanomas (55), and the critical negative regulator of PI3 kinase activity, the lipid phosphatase PTEN, is mutated or epigenetically silenced in many melanomas (56, 57). The recent development of PI3 kinase inhibitors for clinical testing makes it attractive to consider targeting this pathway in this disease. A total kinome sequencing study in melanoma has recently been published, which suggested for the first time that activating mutations in ErbB4 might be present in a subset of melanomas (58). As functional activity of ErbB4 can be inhibited by the already available kinase inhibitor lapatinib (59), it is attractive to consider testing of lapatinib in melanoma patients bearing ErbB4 mutant tumors. Mutations in c-met also have been reported in a series of melanoma cell lines (60), and the recent development of agents with inhibitory activity against c-met for clinical testing makes a similar hypothesis attractive for this molecule.
Several additional signaling pathways have been reported to be active in subsets of melanomas, and functionally important for melanoma biology, but without pharmacologic agents yet available for pathway inhibition. Expression of stabilized β-catenin has been observed in a major subset of melanomas, and β-catenin has been shown to contribute to melanoma development in mouse models (61). Constitutive phosphorylation of the transcription factor Stat3 also has been identified in a subset of melanomas (62). In vitro, knockdown of Stat3 has direct anti-tumor activity, but also induces expression of important immunoregulatory genes, including a subset of chemokines that might mediate lymphocyte trafficking (63). Therefore, Stat3 inhibitors, if developed, might have two complementary mechanisms of action, and synergy of such agents with other immunotherapeutic agents might be anticipated. Developing novel strategies to inhibit the β-catenin and Stat3 pathways in melanoma therefore should receive significant attention.
The very near future of melanoma therapy
Melanoma has recently earned the designation as “an unlikely poster child for personalized cancer therapy” (64). It is not difficult to envision that within the next several years, melanoma tumors will be routinely screened for the presence of a panel of specific markers to determine assignment of individual patients to the most appropriate therapeutic approaches (Table 1). Indeed, analysis of these markers is now frequently used in academic centers for the majority of new patients presenting with metastatic disease and is on the verge of becoming standard practice. Mutations in B-Raf or c-kit will determine eligibility for treatment with the respective specific kinase inhibitors. The activation status of other signaling pathways may be used to predict benefit to other new targeted agents. Expression of selected tumor antigens (e.g. MAGE-3) will be utilized to identify patients for treatment with antigen-specific vaccines. The presence of the “inflamed” tumor microenvironment, anticipated to be characterized using a small gene set analyzed by qRT-PCR much like the OncotypeDx in breast cancer (65), might be utilized to consider patients for a range of immunotherapeutic interventions. Having these predictive biomarkers in hand will affect the care of melanoma patients in multiple ways. First, it should lead to a greater likelihood of clinical response with the first therapeutic modality selected for a given patient. This should in turn lead to improved overall survival of this traditionally difficult to treat population. Second, having a specific molecular pathway that is being targeted should enable identification of resistance or escape mechanisms that, when studied, may lead to the more rapid identification of new therapeutic interventions to pursue in order to maximize clinical benefit. Third, characterization of tumors that lack any of these potential predictive biomarkers (e.g. B-Raf and c-kit wildtype, absence of inflammatory signature) should proceed at an accelerated pace, to identify new pathways that might be amenable to novel therapeutic approaches. Finally, an infrastructure will need to be developed for rapid assessment of patients’ tumors for expression of these predictive biomarkers, using quality-controlled and validated assays and with a rapid turn-around time performed in appropriately credentialed laboratories. This will likely involve a combination of molecular diagnostic laboratories at academic oncology centers and commercial laboratories with expertise in specific assay systems, as well as educational programs for updating community oncologists on the rapidly evolving standard practice. Indeed, the future of melanoma therapy has never looked brighter.
Table 1.
Emerging molecular markers that may facilitate patient-specific therapy in melanoma.
Molecular Biomarker | Therapeutic Modality |
---|---|
Ongoing in development | |
B-Raf V600E | PLX4032 |
Mutant c-kit | Imatinib, nilotinib |
MAGE-3+ | MAGE-3 protein vaccine |
“Inflamed” tumor microenvironment | Various immunotherapeutics |
Future potential | |
Active Notch | GSIs |
Mutant PI3K/PTEN loss | PI3K inhibitors |
Mutant c-met | c-met inhibitors |
Mutant ErbB4 | lapatinib |
Stabilized β-catenin | ? |
Active Stat3 | ? |
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Financial disclosures: The author reports honoraria and clinical trial support from BMS and from GSK-Bio, as well as clinical trial support from Novartis, Roche, Incyte, and Eisai.
References
- 1.Richards JM, Gilewski TA, Ramming K, Mitchel B, Doane LL, Vogelzang NJ. Effective chemotherapy for melanoma after treatment with interleukin-2. Cancer. 1992;69(2):427–9. doi: 10.1002/1097-0142(19920115)69:2<427::aid-cncr2820690225>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
- 2.Atkins MB, Hsu J, Lee S, et al. Phase III trial comparing concurrent biochemotherapy with cisplatin, vinblastine, dacarbazine, interleukin-2, and interferon alfa-2b with cisplatin, vinblastine, and dacarbazine alone in patients with metastatic malignant melanoma (E3695): a trial coordinated by the Eastern Cooperative Oncology Group. J Clin Oncol. 2008;26(35):5748–54. doi: 10.1200/JCO.2008.17.5448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic alterations in melanoma. N Engl J Med. 2005;353(20):2135–47. doi: 10.1056/NEJMoa050092. [DOI] [PubMed] [Google Scholar]
- 4.Coulie PG, Karanikas V, Colau D, et al. A monoclonal cytolytic T-lymphocyte response observed in a melanoma patient vaccinated with a tumor-specific antigenic peptide encoded by gene MAGE-3. Proc Natl Acad Sci U S A. 2001;21:21. doi: 10.1073/pnas.161260098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rosenberg SA, Sherry RM, Morton KE, et al. Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. J Immunol. 2005;175(9):6169–76. doi: 10.4049/jimmunol.175.9.6169. [DOI] [PubMed] [Google Scholar]
- 6.Gajewski TF, Meng Y, Harlin H. Chemokines expressed in melanoma metastases associated with T cell infiltration. Journal of Clinical Oncology; 2007 ASCO Annual Meeting Proceedings Part I.2007. p. 8501. [Google Scholar]
- 7.Gajewski TF, Louahed J, Brichard VG. Gene signature in melanoma associated with clinical activity: a potential clue to unlock cancer immunotherapy. Cancer J. 16(4):399–403. doi: 10.1097/PPO.0b013e3181eacbd8. [DOI] [PubMed] [Google Scholar]
- 8.Harlin H, Meng Y, Peterson AC, et al. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69(7):3077–85. doi: 10.1158/0008-5472.CAN-08-2281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gajewski TF, Zha Y, Thurner B, Schuler G. Association of gene expression profile in melanoma and survival to a dendritic cell-based vaccine. J Clin Oncol. 2009;27(15s):9002. [Google Scholar]
- 10.Vansteenkiste JF, Zielinski M, Dahabreh IJ, et al. Association of gene expression signature and clinical efficacy of MAGE-A3 antigen-specific cancer immunotherapeutic (ASCI) as adjuvant therapy in resected stage IB/II non-small cell lung cancer (NSCLC) J Clin Oncol. 2008 May 20;26(suppl) Abstract 7501. [Google Scholar]
- 11.Harlin H, Kuna TV, Peterson AC, Meng Y, Gajewski TF. Tumor progression despite massive influx of activated CD8(+) T cells in a patient with malignant melanoma ascites. Cancer Immunol Immunother. 2006;55(10):1185–97. doi: 10.1007/s00262-005-0118-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zippelius A, Batard P, Rubio-Godoy V, et al. Effector function of human tumor-specific CD8 T cells in melanoma lesions: a state of local functional tolerance. Cancer Res. 2004;64(8):2865–73. doi: 10.1158/0008-5472.can-03-3066. [DOI] [PubMed] [Google Scholar]
- 13.Appay V, Jandus C, Voelter V, et al. New generation vaccine induces effective melanoma-specific CD8+ T cells in the circulation but not in the tumor site. J Immunol. 2006;177(3):1670–8. doi: 10.4049/jimmunol.177.3.1670. [DOI] [PubMed] [Google Scholar]
- 14.Mortarini R, Piris A, Maurichi A, et al. Lack of terminally differentiated tumor-specific CD8+ T cells at tumor site in spite of antitumor immunity to self-antigens in human metastatic melanoma. Cancer Res. 2003;63(10):2535–45. [PubMed] [Google Scholar]
- 15.Gajewski TF. Failure at the effector phase: immune barriers at the level of the melanoma tumor microenvironment. Clin Cancer Res. 2007;13(18 Pt 1):5256–61. doi: 10.1158/1078-0432.CCR-07-0892. [DOI] [PubMed] [Google Scholar]
- 16.Munn DH, Zhou M, Attwood JT, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281(5380):1191–3. doi: 10.1126/science.281.5380.1191. [DOI] [PubMed] [Google Scholar]
- 17.Dong H, Chen L. B7-H1 pathway and its role in the evasion of tumor immunity. J Mol Med. 2003;81(5):281–7. doi: 10.1007/s00109-003-0430-2. [DOI] [PubMed] [Google Scholar]
- 18.Nomura T, Sakaguchi S. Naturally arising CD25+CD4+ regulatory T cells in tumor immunity. Curr Top Microbiol Immunol. 2005;293:287–302. doi: 10.1007/3-540-27702-1_13. [DOI] [PubMed] [Google Scholar]
- 19.Schwartz RH. T cell anergy. Annu Rev Immunol. 2003;21:305–34. doi: 10.1146/annurev.immunol.21.120601.141110. [DOI] [PubMed] [Google Scholar]
- 20.Brichard VG, Lejeune D. Cancer immunotherapy targeting tumour-specific antigens: towards a new therapy for minimal residual disease. Expert Opin Biol Ther. 2008;8(7):951–68. doi: 10.1517/14712598.8.7.951. [DOI] [PubMed] [Google Scholar]
- 21.Muller AJ, Malachowski WP, Prendergast GC. Indoleamine 2,3-dioxygenase in cancer: targeting pathological immune tolerance with small-molecule inhibitors. Expert Opin Ther Targets. 2005;9(4):831–49. doi: 10.1517/14728222.9.4.831. [DOI] [PubMed] [Google Scholar]
- 22.Uyttenhove C, Pilotte L, Theate I, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med. 2003;9(10):1269–74. doi: 10.1038/nm934. [DOI] [PubMed] [Google Scholar]
- 23.Blank C, Brown I, Peterson AC, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells. Cancer Res. 2004;64(3):1140–5. doi: 10.1158/0008-5472.can-03-3259. [DOI] [PubMed] [Google Scholar]
- 24.Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009;114(8):1545–52. doi: 10.1182/blood-2009-03-206672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Sutmuller RP, van Duivenvoorde LM, van Elsas A, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25(+) regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med. 2001;194(6):823–32. doi: 10.1084/jem.194.6.823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Brown IE, Blank C, Kline J, Kacha AK, Gajewski TF. Homeostatic proliferation as an isolated variable reverses CD8+ T cell anergy and promotes tumor rejection. J Immunol. 2006;177(7):4521–9. doi: 10.4049/jimmunol.177.7.4521. [DOI] [PubMed] [Google Scholar]
- 27.Liu X, Shin N, Koblish HK, et al. Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. Blood. 115(17):3520–30. doi: 10.1182/blood-2009-09-246124. [DOI] [PubMed] [Google Scholar]
- 28.Sznol M, Powderly JD, Smith DC, et al. Safety and antitumor activity of biweekly MDX-1106 (Anti-PD-1, BMS-936558/ONO-4538) in patients with advanced refractory malignancies. J Clin Oncol. 2010;28(15s) Abstract 2506. [Google Scholar]
- 29.Dannull J, Su Z, Rizzieri D, et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest. 2005;115(12):3623–33. doi: 10.1172/JCI25947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rech AJ, Vonderheide RH. Clinical use of anti-CD25 antibody daclizumab to enhance immune responses to tumor antigen vaccination by targeting regulatory T cells. Ann N Y Acad Sci. 2009;1174:99–106. doi: 10.1111/j.1749-6632.2009.04939.x. [DOI] [PubMed] [Google Scholar]
- 31.Rasku MA, Clem AL, Telang S, et al. Transient T cell depletion causes regression of melanoma metastases. J Transl Med. 2008;6:12. doi: 10.1186/1479-5876-6-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rosenberg SA, Dudley ME. Cancer regression in patients with metastatic melanoma after the transfer of autologous antitumor lymphocytes. Proc Natl Acad Sci U S A. 2004;101(Suppl 2):14639–45. doi: 10.1073/pnas.0405730101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sullivan RJ, Hoshida Y, Brunet J, et al. A single center experience with high-dose IL-2 treatment for patients with advanced melanoma and pilot investigation of a novel gene expression signature as a predictor of response. J Clin Oncol. 2009;27(15S) Abstract 9003. [Google Scholar]
- 34.Hamid O, Chasalow SD, Tsuchihashi Z, Alaparthy S, Galbraith S, Berman D. Association of baseline and on-study tumor biopsy markers with clinical activity in patients with advanced melanoma treated with ipilimumab. J Clin Oncol. 2009;27(15s) Abstract 9008. [Google Scholar]
- 35.Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949–54. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
- 36.Eisen T, Ahmad T, Flaherty KT, et al. Sorafenib in advanced melanoma: a Phase II randomised discontinuation trial analysis. Br J Cancer. 2006;95(5):581–6. doi: 10.1038/sj.bjc.6603291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Flaherty KT, Schiller J, Schuchter LM, et al. A phase I trial of the oral, multikinase inhibitor sorafenib in combination with carboplatin and paclitaxel. Clin Cancer Res. 2008;14(15):4836–42. doi: 10.1158/1078-0432.CCR-07-4123. [DOI] [PubMed] [Google Scholar]
- 38.Flaherty KT, Lee SJ, Schuchter LM, et al. Final results of E2603: A double-blind, randomized phase III trial comparing carboplatin ©/paclitaxel (P) with or without sorafenib (S) in metastatic melanoma. Clin Oncol. 2010;28(15s) Abstract 8511. [Google Scholar]
- 39.Gajewski TF, Niedzwiecki D, Johnson J, et al. Phase II study of the farnesyltransferase inhibitor R115777 in advanced melanoma: CALGB 500104. J Clin Oncol. 2006;24(18s) doi: 10.1186/1479-5876-10-246. Abstract 8014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 363(9):809–19. doi: 10.1056/NEJMoa1002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene mutations and copy number in melanoma subtypes. Clin Cancer Res. 2008;14(21):6821–8. doi: 10.1158/1078-0432.CCR-08-0575. [DOI] [PubMed] [Google Scholar]
- 42.Satzger I, Schaefer T, Kuettler U, et al. Analysis of c-KIT expression and KIT gene mutation in human mucosal melanomas. Br J Cancer. 2008;99(12):2065–9. doi: 10.1038/sj.bjc.6604791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Heinrich MC, Corless CL, Demetri GD, et al. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. J Clin Oncol. 2003;21(23):4342–9. doi: 10.1200/JCO.2003.04.190. [DOI] [PubMed] [Google Scholar]
- 44.Hodi FS, Friedlander P, Corless CL, et al. Major response to imatinib mesylate in KIT-mutated melanoma. J Clin Oncol. 2008;26(12):2046–51. doi: 10.1200/JCO.2007.14.0707. [DOI] [PubMed] [Google Scholar]
- 45.Lutzky J, Bauer J, Bastian BC. Dose-dependent, complete response to imatinib of a metastatic mucosal melanoma with a K642E KIT mutation. Pigment Cell Melanoma Res. 2008;21(4):492–3. doi: 10.1111/j.1755-148X.2008.00475.x. [DOI] [PubMed] [Google Scholar]
- 46.Quintas-Cardama A, Lazar AJ, Woodman SE, Kim K, Ross M, Hwu P. Complete response of stage IV anal mucosal melanoma expressing KIT Val560Asp to the multikinase inhibitor sorafenib. Nat Clin Pract Oncol. 2008;5(12):737–40. doi: 10.1038/ncponc1251. [DOI] [PubMed] [Google Scholar]
- 47.Satzger I, Kuttler U, Volker B, Schenck F, Kapp A, Gutzmer R. Anal mucosal melanoma with KIT-activating mutation and response to imatinib therapy--case report and review of the literature. Dermatology. 220(1):77–81. doi: 10.1159/000265558. [DOI] [PubMed] [Google Scholar]
- 48.Wyman K, Atkins MB, Prieto V, et al. Multicenter Phase II trial of high-dose imatinib mesylate in metastatic melanoma: significant toxicity with no clinical efficacy. Cancer. 2006;106(9):2005–11. doi: 10.1002/cncr.21834. [DOI] [PubMed] [Google Scholar]
- 49.Kim KB, Eton O, Davis DW, et al. Phase II trial of imatinib mesylate in patients with metastatic melanoma. Br J Cancer. 2008;99(5):734–40. doi: 10.1038/sj.bjc.6604482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352(10):997–1003. doi: 10.1056/NEJMoa043331. [DOI] [PubMed] [Google Scholar]
- 51.Tawby HA, Buch S, Pancoska P, et al. Prediction of response to alkylator-based chemotherapy in metastatic melanoma (MM) using gene expression and promoter methylation signatures. J Clin Oncol. 2010;27(15s) Abstract 9009. [Google Scholar]
- 52.Gajewski TF, Sosman J, Gerson SL, et al. Phase II trial of the O6-alkylguanine DNA alkyltransferase inhibitor O6-benzylguanine and 1,3-bis(2-chloroethyl)-1-nitrosourea in advanced melanoma. Clin Cancer Res. 2005;11(21):7861–5. doi: 10.1158/1078-0432.CCR-05-0060. [DOI] [PubMed] [Google Scholar]
- 53.Massi D, Tarantini F, Franchi A, et al. Evidence for differential expression of Notch receptors and their ligands in melanocytic nevi and cutaneous malignant melanoma. Mod Pathol. 2006;19(2):246–54. doi: 10.1038/modpathol.3800526. [DOI] [PubMed] [Google Scholar]
- 54.Tolcher AW, Mikulski SM, Messersmith WA, et al. A phase I study of RO4929097, a novel gamma secretase inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2010;28(15s) doi: 10.1200/JCO.2011.36.8282. Abstract 2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Curtin JA, Stark MS, Pinkel D, Hayward NK, Bastian BC. PI3-kinase subunits are infrequent somatic targets in melanoma. J Invest Dermatol. 2006;126(7):1660–3. doi: 10.1038/sj.jid.5700311. [DOI] [PubMed] [Google Scholar]
- 56.Zhou XP, Gimm O, Hampel H, Niemann T, Walker MJ, Eng C. Epigenetic PTEN silencing in malignant melanomas without PTEN mutation. Am J Pathol. 2000;157(4):1123–8. doi: 10.1016/S0002-9440(10)64627-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Celebi JT, Shendrik I, Silvers DN, Peacocke M. Identification of PTEN mutations in metastatic melanoma specimens. J Med Genet. 2000;37(9):653–7. doi: 10.1136/jmg.37.9.653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Prickett TD, Agrawal NS, Wei X, et al. Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4. Nat Genet. 2009;41(10):1127–32. doi: 10.1038/ng.438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Bilancia D, Rosati G, Dinota A, Germano D, Romano R, Manzione L. Lapatinib in breast cancer. Ann Oncol. 2007;18(Suppl 6):vi26–30. doi: 10.1093/annonc/mdm220. [DOI] [PubMed] [Google Scholar]
- 60.Puri N, Ahmed S, Janamanchi V, et al. c-Met is a potentially new therapeutic target for treatment of human melanoma. Clin Cancer Res. 2007;13(7):2246–53. doi: 10.1158/1078-0432.CCR-06-0776. [DOI] [PubMed] [Google Scholar]
- 61.Larue L, Delmas V. The WNT/Beta-catenin pathway in melanoma. Front Biosci. 2006;11:733–42. doi: 10.2741/1831. [DOI] [PubMed] [Google Scholar]
- 62.Messina JL, Yu H, Riker AI, Munster PN, Jove RL, Daud AI. Activated stat-3 in melanoma. Cancer Control. 2008;15(3):196–201. doi: 10.1177/107327480801500302. [DOI] [PubMed] [Google Scholar]
- 63.Burdelya L, Kujawski M, Niu G, et al. Stat3 activity in melanoma cells affects migration of immune effector cells and nitric oxide-mediated antitumor effects. J Immunol. 2005;174(7):3925–31. doi: 10.4049/jimmunol.174.7.3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Smalley KS, Sondak VK. Melanoma--an unlikely poster child for personalized cancer therapy. N Engl J Med. 363(9):876–8. doi: 10.1056/NEJMe1005370. [DOI] [PubMed] [Google Scholar]
- 65.Kim C, Paik S. Gene-expression-based prognostic assays for breast cancer. Nat Rev Clin Oncol. 7(6):340–7. doi: 10.1038/nrclinonc.2010.61. [DOI] [PubMed] [Google Scholar]