Skip to main content
Top
Published in: Clinical & Experimental Metastasis 5-6/2018

01-08-2018 | Review

Review of diagnostic, prognostic, and predictive biomarkers in melanoma

Authors: Jacob S. Ankeny, Brian Labadie, Jason Luke, Eddy Hsueh, Jane Messina, Jonathan S. Zager

Published in: Clinical & Experimental Metastasis | Issue 5-6/2018

Login to get access

Abstract

Melanoma is an aggressive cutaneous malignancy with rapidly rising incidence. Diagnosis of controversial melanocytic lesions, correct prognostication of patients, selection of appropriate adjuvant and systemic therapies, and prediction of response to a given therapy remain very real challenges. Despite these challenges, multiple high throughput, nucleic-acid based biomarkers have been developed that can be assayed from histologic tissue specimens. FISH, CGH, Decision-Dx, and other multi-marker assays have been combined to improve overall predictability. This review discusses some of the most promising nucleic acid based assays that can be obtained from tissue specimens to assist with diagnosis, prognostication, and prediction of treatment response.
Literature
1.
go back to reference Siegel RL, Miller KD, Jemal A (2017) Cancer statistics. CA Cancer J Clin 67(1):7–30CrossRef Siegel RL, Miller KD, Jemal A (2017) Cancer statistics. CA Cancer J Clin 67(1):7–30CrossRef
2.
go back to reference Gimotty PA, Guerry D (2010) Prognostication in thin cutaneous melanomas. Arch Pathol Lab Med 134(12):1758–1763PubMed Gimotty PA, Guerry D (2010) Prognostication in thin cutaneous melanomas. Arch Pathol Lab Med 134(12):1758–1763PubMed
3.
go back to reference Whiteman DC, Baade PD, Olsen CM (2015) More people die from thin melanomas (1 mm) than from thick melanomas (> 4 mm) in Queensland, Australia. J Invest Dermatol 135(4):1190–1193CrossRef Whiteman DC, Baade PD, Olsen CM (2015) More people die from thin melanomas (1 mm) than from thick melanomas (> 4 mm) in Queensland, Australia. J Invest Dermatol 135(4):1190–1193CrossRef
4.
go back to reference Wong SL et al (2005) A nomogram that predicts the presence of sentinel node metastasis in melanoma with better discrimination than the American Joint Committee on Cancer staging system. Ann Surg Oncol 12(4):282–288CrossRef Wong SL et al (2005) A nomogram that predicts the presence of sentinel node metastasis in melanoma with better discrimination than the American Joint Committee on Cancer staging system. Ann Surg Oncol 12(4):282–288CrossRef
5.
go back to reference Mahar AL, McShane LM, Groome PA, Compton CC (2013) A survey of clinical prediction tools in colorectal and lung cancers and melanoma. J Clin Oncol 31(15S): 1592 Mahar AL, McShane LM, Groome PA, Compton CC (2013) A survey of clinical prediction tools in colorectal and lung cancers and melanoma. J Clin Oncol 31(15S): 1592
6.
go back to reference Mahar AL et al (2016) Critical assessment of clinical prognostic tools in melanoma. Ann Surg Oncol 23(9):2753–2761CrossRef Mahar AL et al (2016) Critical assessment of clinical prognostic tools in melanoma. Ann Surg Oncol 23(9):2753–2761CrossRef
7.
go back to reference Gould Rothberg BE, Bracken MB, Rimm DL (2009) Tissue biomarkers for prognosis in cutaneous melanoma: a systematic review and meta-analysis. J Natl Cancer Inst 101(7):452–474CrossRef Gould Rothberg BE, Bracken MB, Rimm DL (2009) Tissue biomarkers for prognosis in cutaneous melanoma: a systematic review and meta-analysis. J Natl Cancer Inst 101(7):452–474CrossRef
8.
go back to reference Schramm SJ, Mann GJ (2011) Melanoma prognosis: a REMARK-based systematic review and bioinformatic analysis of immunohistochemical and gene microarray studies. Mol Cancer Ther 10(8):1520–1528CrossRef Schramm SJ, Mann GJ (2011) Melanoma prognosis: a REMARK-based systematic review and bioinformatic analysis of immunohistochemical and gene microarray studies. Mol Cancer Ther 10(8):1520–1528CrossRef
9.
go back to reference Gould Rothberg BE et al (2009) Melanoma prognostic model using tissue microarrays and genetic algorithms. J Clin Oncol 27(34):5772–5780CrossRef Gould Rothberg BE et al (2009) Melanoma prognostic model using tissue microarrays and genetic algorithms. J Clin Oncol 27(34):5772–5780CrossRef
10.
go back to reference Piras F et al (2008) Combinations of apoptosis and cell-cycle control biomarkers predict the outcome of human melanoma. Oncol Rep 20(2):271–277PubMed Piras F et al (2008) Combinations of apoptosis and cell-cycle control biomarkers predict the outcome of human melanoma. Oncol Rep 20(2):271–277PubMed
11.
go back to reference Kashani-Sabet M et al (2009) A multimarker prognostic assay for primary cutaneous melanoma. Clin Cancer Res 15(22):6987–6992CrossRef Kashani-Sabet M et al (2009) A multimarker prognostic assay for primary cutaneous melanoma. Clin Cancer Res 15(22):6987–6992CrossRef
12.
go back to reference Gerami P et al (2015) Development of a prognostic genetic signature to predict the metastatic risk associated with cutaneous melanoma. Clin Cancer Res 21(1):175–183CrossRef Gerami P et al (2015) Development of a prognostic genetic signature to predict the metastatic risk associated with cutaneous melanoma. Clin Cancer Res 21(1):175–183CrossRef
13.
go back to reference Gerami P et al (2015) Gene expression profiling for molecular staging of cutaneous melanoma in patients undergoing sentinel lymph node biopsy. J Am Acad Dermatol 72(5):780–785.e3CrossRef Gerami P et al (2015) Gene expression profiling for molecular staging of cutaneous melanoma in patients undergoing sentinel lymph node biopsy. J Am Acad Dermatol 72(5):780–785.e3CrossRef
14.
go back to reference Gerami P et al (2009) Fluorescence in situ hybridization (FISH) as an ancillary diagnostic tool in the diagnosis of melanoma. Am J Surg Pathol 33(8):1146–1156CrossRef Gerami P et al (2009) Fluorescence in situ hybridization (FISH) as an ancillary diagnostic tool in the diagnosis of melanoma. Am J Surg Pathol 33(8):1146–1156CrossRef
15.
go back to reference Scolyer RA et al (2010) Histologically ambiguous (“borderline”) primary cutaneous melanocytic tumors: approaches to patient management including the roles of molecular testing and sentinel lymph node biopsy. Arch Pathol Lab Med 134(12):1770–1777PubMed Scolyer RA et al (2010) Histologically ambiguous (“borderline”) primary cutaneous melanocytic tumors: approaches to patient management including the roles of molecular testing and sentinel lymph node biopsy. Arch Pathol Lab Med 134(12):1770–1777PubMed
16.
go back to reference Dalton SR et al (2010) Use of fluorescence in situ hybridization (FISH) to distinguish intranodal nevus from metastatic melanoma. Am J Surg Pathol 34(2):231–237CrossRef Dalton SR et al (2010) Use of fluorescence in situ hybridization (FISH) to distinguish intranodal nevus from metastatic melanoma. Am J Surg Pathol 34(2):231–237CrossRef
17.
go back to reference Pouryazdanparast P et al (2009) Distinguishing epithelioid blue nevus from blue nevus-like cutaneous melanoma metastasis using fluorescence in situ hybridization. Am J Surg Pathol 33(9):1396–1400CrossRef Pouryazdanparast P et al (2009) Distinguishing epithelioid blue nevus from blue nevus-like cutaneous melanoma metastasis using fluorescence in situ hybridization. Am J Surg Pathol 33(9):1396–1400CrossRef
18.
go back to reference Gerami P et al (2009) Fluorescence in situ hybridization for distinguishing nevoid melanomas from mitotically active nevi. Am J Surg Pathol 33(12):1783–1788CrossRef Gerami P et al (2009) Fluorescence in situ hybridization for distinguishing nevoid melanomas from mitotically active nevi. Am J Surg Pathol 33(12):1783–1788CrossRef
19.
go back to reference Gerami P et al (2012) A highly specific and discriminatory FISH assay for distinguishing between benign and malignant melanocytic neoplasms. Am J Surg Pathol 36(6):808–817CrossRef Gerami P et al (2012) A highly specific and discriminatory FISH assay for distinguishing between benign and malignant melanocytic neoplasms. Am J Surg Pathol 36(6):808–817CrossRef
20.
go back to reference Gaiser T et al (2010) Classifying ambiguous melanocytic lesions with FISH and correlation with clinical long-term follow up. Mod Pathol 23(3):413–419CrossRef Gaiser T et al (2010) Classifying ambiguous melanocytic lesions with FISH and correlation with clinical long-term follow up. Mod Pathol 23(3):413–419CrossRef
21.
go back to reference Massi D et al (2011) Atypical Spitzoid melanocytic tumors: a morphological, mutational, and FISH analysis. J Am Acad Dermatol 64(5):919–935CrossRef Massi D et al (2011) Atypical Spitzoid melanocytic tumors: a morphological, mutational, and FISH analysis. J Am Acad Dermatol 64(5):919–935CrossRef
22.
go back to reference Tetzlaff MT et al (2013) Ambiguous melanocytic tumors in a tertiary referral center: the contribution of fluorescence in situ hybridization (FISH) to conventional histopathologic and immunophenotypic analyses. Am J Surg Pathol 37(12):1783–1796CrossRef Tetzlaff MT et al (2013) Ambiguous melanocytic tumors in a tertiary referral center: the contribution of fluorescence in situ hybridization (FISH) to conventional histopathologic and immunophenotypic analyses. Am J Surg Pathol 37(12):1783–1796CrossRef
23.
go back to reference Vergier B et al (2011) Fluorescence in situ hybridization, a diagnostic aid in ambiguous melanocytic tumors: European study of 113 cases. Mod Pathol 24(5):613–623CrossRef Vergier B et al (2011) Fluorescence in situ hybridization, a diagnostic aid in ambiguous melanocytic tumors: European study of 113 cases. Mod Pathol 24(5):613–623CrossRef
24.
go back to reference Bauer J, Bastian BC (2006) Distinguishing melanocytic nevi from melanoma by DNA copy number changes: comparative genomic hybridization as a research and diagnostic tool. Dermatol Ther 19(1):40–49CrossRef Bauer J, Bastian BC (2006) Distinguishing melanocytic nevi from melanoma by DNA copy number changes: comparative genomic hybridization as a research and diagnostic tool. Dermatol Ther 19(1):40–49CrossRef
25.
go back to reference Bastian BC et al (1998) Chromosomal gains and losses in primary cutaneous melanomas detected by comparative genomic hybridization. Cancer Res 58(10):2170–2175 Bastian BC et al (1998) Chromosomal gains and losses in primary cutaneous melanomas detected by comparative genomic hybridization. Cancer Res 58(10):2170–2175
26.
go back to reference Ali L et al. (2010) Correlating array comparative genomic hybridization findings with histology and outcome in spitzoid melanocytic neoplasms. Int J Clin Exp Pathol 3(6): 593–599 Ali L et al. (2010) Correlating array comparative genomic hybridization findings with histology and outcome in spitzoid melanocytic neoplasms. Int J Clin Exp Pathol 3(6): 593–599
27.
go back to reference Bastian BC et al (2003) Classifying melanocytic tumors based on DNA copy number changes. Am J Pathol 163(5):1765–1770CrossRef Bastian BC et al (2003) Classifying melanocytic tumors based on DNA copy number changes. Am J Pathol 163(5):1765–1770CrossRef
28.
go back to reference Clarke LE et al (2015) Clinical validation of a gene expression signature that differentiates benign nevi from malignant melanoma. J Cutan Pathol 42(4):244–252CrossRef Clarke LE et al (2015) Clinical validation of a gene expression signature that differentiates benign nevi from malignant melanoma. J Cutan Pathol 42(4):244–252CrossRef
29.
go back to reference Minca EC et al (2016) Comparison between melanoma gene expression score and fluorescence in situ hybridization for the classification of melanocytic lesions. Mod Pathol 29(8):832–843CrossRef Minca EC et al (2016) Comparison between melanoma gene expression score and fluorescence in situ hybridization for the classification of melanocytic lesions. Mod Pathol 29(8):832–843CrossRef
30.
go back to reference Network NCC (2018) NCCN Guidelines Version 2.2018, in NCCN Clnical Practice Guidelines in Oncology. Coit DG (ed) National Comprehensive Cancer Network Network NCC (2018) NCCN Guidelines Version 2.2018, in NCCN Clnical Practice Guidelines in Oncology. Coit DG (ed) National Comprehensive Cancer Network
31.
go back to reference Abbas O, Miller DD, Bhawan J (2014) Cutaneous malignant melanoma: update on diagnostic and prognostic biomarkers. Am J Dermatopathol 36(5):363–379CrossRef Abbas O, Miller DD, Bhawan J (2014) Cutaneous malignant melanoma: update on diagnostic and prognostic biomarkers. Am J Dermatopathol 36(5):363–379CrossRef
32.
go back to reference Adler NR et al (2017) Metastatic pathways in patients with cutaneous melanoma. Pigment Cell Melanoma Res 30(1):13–27CrossRef Adler NR et al (2017) Metastatic pathways in patients with cutaneous melanoma. Pigment Cell Melanoma Res 30(1):13–27CrossRef
33.
go back to reference Nodin B et al (2012) High MCM3 expression is an independent biomarker of poor prognosis and correlates with reduced RBM3 expression in a prospective cohort of malignant melanoma. Diagn Pathol 7:82CrossRef Nodin B et al (2012) High MCM3 expression is an independent biomarker of poor prognosis and correlates with reduced RBM3 expression in a prospective cohort of malignant melanoma. Diagn Pathol 7:82CrossRef
34.
go back to reference Nielsen PS et al (2013) Proliferation indices of phosphohistone H3 and Ki67: strong prognostic markers in a consecutive cohort with stage I/II melanoma. Mod Pathol 26(3):404–413CrossRef Nielsen PS et al (2013) Proliferation indices of phosphohistone H3 and Ki67: strong prognostic markers in a consecutive cohort with stage I/II melanoma. Mod Pathol 26(3):404–413CrossRef
35.
go back to reference Donizy P et al (2016) Golgi-related proteins GOLPH2 (GP73/GOLM1) and GOLPH3 (GOPP1/MIDAS) in cutaneous melanoma: patterns of expression and prognostic significance. Int J Mol Sci 17(10):1619CrossRef Donizy P et al (2016) Golgi-related proteins GOLPH2 (GP73/GOLM1) and GOLPH3 (GOPP1/MIDAS) in cutaneous melanoma: patterns of expression and prognostic significance. Int J Mol Sci 17(10):1619CrossRef
36.
go back to reference Fohn LE et al (2011) D2-40 lymphatic marker for detecting lymphatic invasion in thin to intermediate thickness melanomas: association with sentinel lymph node status and prognostic value-a retrospective case study. J Am Acad Dermatol 64(2):336–345CrossRef Fohn LE et al (2011) D2-40 lymphatic marker for detecting lymphatic invasion in thin to intermediate thickness melanomas: association with sentinel lymph node status and prognostic value-a retrospective case study. J Am Acad Dermatol 64(2):336–345CrossRef
37.
go back to reference Han D et al (2013) Clinicopathologic predictors of sentinel lymph node metastasis in thin melanoma. J Clin Oncol 31(35):4387–4393CrossRef Han D et al (2013) Clinicopathologic predictors of sentinel lymph node metastasis in thin melanoma. J Clin Oncol 31(35):4387–4393CrossRef
38.
go back to reference Rangel J et al (2008) Osteopontin as a molecular prognostic marker for melanoma. Cancer 112(1):144–150CrossRef Rangel J et al (2008) Osteopontin as a molecular prognostic marker for melanoma. Cancer 112(1):144–150CrossRef
39.
go back to reference Rangel J et al (2008) Novel role for RGS1 in melanoma progression. Am J Surg Pathol 32(8):1207–1212CrossRef Rangel J et al (2008) Novel role for RGS1 in melanoma progression. Am J Surg Pathol 32(8):1207–1212CrossRef
40.
go back to reference Leiter U et al (2016) Complete lymph node dissection versus no dissection in patients with sentinel lymph node biopsy positive melanoma (DeCOG-SLT): a multicentre, randomised, phase 3 trial. Lancet Oncol 17(6):757–767CrossRef Leiter U et al (2016) Complete lymph node dissection versus no dissection in patients with sentinel lymph node biopsy positive melanoma (DeCOG-SLT): a multicentre, randomised, phase 3 trial. Lancet Oncol 17(6):757–767CrossRef
41.
go back to reference Faries MB et al (2017) Completion dissection or observation for sentinel-node metastasis in melanoma. N Engl J Med 376(23):2211–2222CrossRef Faries MB et al (2017) Completion dissection or observation for sentinel-node metastasis in melanoma. N Engl J Med 376(23):2211–2222CrossRef
42.
go back to reference Damude S et al (2016) The predictive power of serum S-100B for non-sentinel node positivity in melanoma patients. Eur J Surg Oncol 42(4):545–551CrossRef Damude S et al (2016) The predictive power of serum S-100B for non-sentinel node positivity in melanoma patients. Eur J Surg Oncol 42(4):545–551CrossRef
43.
go back to reference Wevers KP et al (2013) Assessment of a new scoring system for predicting non-sentinel node positivity in sentinel node-positive melanoma patients. Eur J Surg Oncol 39(2):179–184CrossRef Wevers KP et al (2013) Assessment of a new scoring system for predicting non-sentinel node positivity in sentinel node-positive melanoma patients. Eur J Surg Oncol 39(2):179–184CrossRef
44.
go back to reference van der Ploeg AP et al (2011) Prognosis in patients with sentinel node-positive melanoma is accurately defined by the combined Rotterdam tumor load and Dewar topography criteria. J Clin Oncol 29(16):2206–2214CrossRef van der Ploeg AP et al (2011) Prognosis in patients with sentinel node-positive melanoma is accurately defined by the combined Rotterdam tumor load and Dewar topography criteria. J Clin Oncol 29(16):2206–2214CrossRef
45.
go back to reference Pastushenko I et al (2016) Increased angiogenesis and lymphangiogenesis in metastatic sentinel lymph nodes is associated with nonsentinel lymph node involvement and distant metastasis in patients with melanoma. Am J Dermatopathol 38(5):338–346CrossRef Pastushenko I et al (2016) Increased angiogenesis and lymphangiogenesis in metastatic sentinel lymph nodes is associated with nonsentinel lymph node involvement and distant metastasis in patients with melanoma. Am J Dermatopathol 38(5):338–346CrossRef
46.
go back to reference Veronesi U et al (1988) Thin stage I primary cutaneous malignant melanoma: comparison of excision with margins of 1 or 3 cm. N Engl J Med 318(18):1159–1162CrossRef Veronesi U et al (1988) Thin stage I primary cutaneous malignant melanoma: comparison of excision with margins of 1 or 3 cm. N Engl J Med 318(18):1159–1162CrossRef
47.
go back to reference Ringborg U et al (1996) Resection margins of 2 versus 5 cm for cutaneous malignant melanoma with a tumor thickness of 0.8 to 2.0 mm: randomized study by the Swedish Melanoma Study Group. Cancer 77(9):1809–1814CrossRef Ringborg U et al (1996) Resection margins of 2 versus 5 cm for cutaneous malignant melanoma with a tumor thickness of 0.8 to 2.0 mm: randomized study by the Swedish Melanoma Study Group. Cancer 77(9):1809–1814CrossRef
48.
go back to reference Balch CM et al (2001) Long-term results of a prospective surgical trial comparing 2 cm vs. 4 cm excision margins for 740 patients with 1–4 mm melanomas. Ann Surg Oncol 8(2):101–108PubMed Balch CM et al (2001) Long-term results of a prospective surgical trial comparing 2 cm vs. 4 cm excision margins for 740 patients with 1–4 mm melanomas. Ann Surg Oncol 8(2):101–108PubMed
49.
go back to reference Thomas JM et al (2004) Excision margins in high-risk malignant melanoma. N Engl J Med 350(8):757–766CrossRef Thomas JM et al (2004) Excision margins in high-risk malignant melanoma. N Engl J Med 350(8):757–766CrossRef
50.
go back to reference Gillgren P et al (2011) 2-cm versus 4-cm surgical excision margins for primary cutaneous melanoma thicker than 2 mm: a randomised, multicentre trial. Lancet 378(9803):1635–1642CrossRef Gillgren P et al (2011) 2-cm versus 4-cm surgical excision margins for primary cutaneous melanoma thicker than 2 mm: a randomised, multicentre trial. Lancet 378(9803):1635–1642CrossRef
51.
go back to reference Doepker MP et al (2016) Is a wider margin (2 cm vs. 1 cm) for a 1.01-2.0 mm melanoma necessary? Ann Surg Oncol 23(7):2336–2342CrossRef Doepker MP et al (2016) Is a wider margin (2 cm vs. 1 cm) for a 1.01-2.0 mm melanoma necessary? Ann Surg Oncol 23(7):2336–2342CrossRef
52.
go back to reference Hsueh EC et al (2017) Interim analysis of survival in a prospective, multi-center registry cohort of cutaneous melanoma tested with a prognostic 31-gene expression profile test. J Hematol Oncol 10(1):152CrossRef Hsueh EC et al (2017) Interim analysis of survival in a prospective, multi-center registry cohort of cutaneous melanoma tested with a prognostic 31-gene expression profile test. J Hematol Oncol 10(1):152CrossRef
53.
go back to reference Zager JS et al (2018) Performance of a prognostic 31-gene expression profile in an independent cohort of 523 cutaneous melanoma patients. BMC Cancer 18(1):130CrossRef Zager JS et al (2018) Performance of a prognostic 31-gene expression profile in an independent cohort of 523 cutaneous melanoma patients. BMC Cancer 18(1):130CrossRef
54.
go back to reference Ferris LK et al (2017) Identification of high-risk cutaneous melanoma tumors is improved when combining the online American Joint Committee on Cancer Individualized Melanoma Patient Outcome Prediction Tool with a 31-gene expression profile-based classification. J Am Acad Dermatol 76(5):818–825 e3CrossRef Ferris LK et al (2017) Identification of high-risk cutaneous melanoma tumors is improved when combining the online American Joint Committee on Cancer Individualized Melanoma Patient Outcome Prediction Tool with a 31-gene expression profile-based classification. J Am Acad Dermatol 76(5):818–825 e3CrossRef
55.
go back to reference Eggermont AM et al (2015) Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol 16(5):522–530CrossRef Eggermont AM et al (2015) Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol 16(5):522–530CrossRef
56.
go back to reference Weber J et al (2017) Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med 377(19):1824–1835CrossRef Weber J et al (2017) Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med 377(19):1824–1835CrossRef
57.
go back to reference Long GV et al (2017) Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N Engl J Med 377(19):1813–1823CrossRef Long GV et al (2017) Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N Engl J Med 377(19):1813–1823CrossRef
58.
go back to reference Wolchok JD et al (2017) Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 377(14):1345–1356CrossRef Wolchok JD et al (2017) Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 377(14):1345–1356CrossRef
59.
go back to reference Hodi FS et al (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711–723CrossRef Hodi FS et al (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711–723CrossRef
60.
go back to reference Robert C et al (2015) Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 372(26):2521–2532CrossRef Robert C et al (2015) Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 372(26):2521–2532CrossRef
61.
go back to reference Robert C et al (2015) Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med 372(1):30–39CrossRef Robert C et al (2015) Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med 372(1):30–39CrossRef
62.
go back to reference Larkin J et al (2014) Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med 371(20):1867–1876CrossRef Larkin J et al (2014) Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med 371(20):1867–1876CrossRef
63.
go back to reference Luke JJ et al (2017) Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol 14(8):463–482CrossRef Luke JJ et al (2017) Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol 14(8):463–482CrossRef
64.
go back to reference Daud AI et al (2016) Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody pembrolizumab in melanoma. J Clin Oncol 34(34):4102–4109CrossRef Daud AI et al (2016) Programmed death-ligand 1 expression and response to the anti-programmed death 1 antibody pembrolizumab in melanoma. J Clin Oncol 34(34):4102–4109CrossRef
65.
go back to reference Snyder A et al (2014) Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 371(23):2189–2199CrossRef Snyder A et al (2014) Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 371(23):2189–2199CrossRef
66.
go back to reference Johnson DB et al (2016) Targeted next generation sequencing identifies markers of response to PD-1 blockade. Cancer Immunol Res 4(11):959–967CrossRef Johnson DB et al (2016) Targeted next generation sequencing identifies markers of response to PD-1 blockade. Cancer Immunol Res 4(11):959–967CrossRef
67.
go back to reference Harlin H et al (2009) Chemokine expression in melanoma metastases associated with CD8 + T-cell recruitment. Cancer Res 69(7):3077–3085CrossRef Harlin H et al (2009) Chemokine expression in melanoma metastases associated with CD8 + T-cell recruitment. Cancer Res 69(7):3077–3085CrossRef
68.
go back to reference Ji RR et al (2012) An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother 61(7):1019–1031CrossRef Ji RR et al (2012) An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother 61(7):1019–1031CrossRef
69.
go back to reference Gajewski TF, Schreiber H, Fu YX (2013) Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 14(10):1014–1022CrossRef Gajewski TF, Schreiber H, Fu YX (2013) Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 14(10):1014–1022CrossRef
70.
go back to reference Ayers M et al (2017) IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 127(8):2930–2940CrossRef Ayers M et al (2017) IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 127(8):2930–2940CrossRef
Metadata
Title
Review of diagnostic, prognostic, and predictive biomarkers in melanoma
Authors
Jacob S. Ankeny
Brian Labadie
Jason Luke
Eddy Hsueh
Jane Messina
Jonathan S. Zager
Publication date
01-08-2018
Publisher
Springer Netherlands
Published in
Clinical & Experimental Metastasis / Issue 5-6/2018
Print ISSN: 0262-0898
Electronic ISSN: 1573-7276
DOI
https://doi.org/10.1007/s10585-018-9892-z

Other articles of this Issue 5-6/2018

Clinical & Experimental Metastasis 5-6/2018 Go to the issue

Editorial

Editorial

Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine