Skip to main content
Top
Published in: Clinical & Experimental Metastasis 4/2011

01-04-2011 | Review

Genetic factors in metastatic progression of cutaneous melanoma: the future role of circulating melanoma cells in prognosis and management

Authors: A. Ireland, M. Millward, R. Pearce, M. Lee, M. Ziman

Published in: Clinical & Experimental Metastasis | Issue 4/2011

Login to get access

Abstract

The greatest potential for improvement of outcome for patients with Cutaneous Malignant Melanoma lies in the prevention of systemic metastasis. Despite extensive investigation, current prognostic indicators either alone or in combination, although related to melanoma progression, are not sufficient to accurately predict the pattern of progression and outcome for any individual patient. Metastasis related death has been recorded in patients initially diagnosed with early stage tumour as well as in patients many years after initial tumour removal. The trouble finding a predictable pattern in the puzzle of melanoma progression may be linked to the fact that most of the material studied for prognosis is either, cutaneous primaries or metastatic deposits, rather than the melanoma cells in the circulatory system which are responsible for disease progression. In this review article we discuss the potential use of circulating tumour cell (CTC) detection and quantification for identifying patients at risk of metastatic deposits. We also discuss current therapies for the treatment of metastatic melanoma and analyse how CTCs may be used to evaluate the effectiveness of current therapies and to pinpoint patients who require further treatment.
Literature
1.
go back to reference Lewis TB et al (2005) Molecular classification of melanoma using real-time quantitative reverse transcriptase-polymerase chain reaction. Cancer 104:1678–1686PubMed Lewis TB et al (2005) Molecular classification of melanoma using real-time quantitative reverse transcriptase-polymerase chain reaction. Cancer 104:1678–1686PubMed
3.
go back to reference Jack A et al (2006) The treatment of melanoma with an emphasis on immunotherapeutic strategies. Surg Oncol 15:13–24PubMed Jack A et al (2006) The treatment of melanoma with an emphasis on immunotherapeutic strategies. Surg Oncol 15:13–24PubMed
4.
go back to reference Lee RT et al (2008) Melanoma presenting as circulating tumor cells associated with failed angiogenesis. Melanoma Res 18:289–294PubMed Lee RT et al (2008) Melanoma presenting as circulating tumor cells associated with failed angiogenesis. Melanoma Res 18:289–294PubMed
5.
go back to reference Zbytek B et al (2008) Current concepts of metastasis in melanoma. Expert Rev Dermatol 3:569–585PubMed Zbytek B et al (2008) Current concepts of metastasis in melanoma. Expert Rev Dermatol 3:569–585PubMed
6.
go back to reference Yang AS, Chapman PB (2009) The history and future of chemotherapy for melanoma. Hematol Oncol Clin North Am 23:583–597PubMed Yang AS, Chapman PB (2009) The history and future of chemotherapy for melanoma. Hematol Oncol Clin North Am 23:583–597PubMed
7.
go back to reference Breslow A (1975) Tumor thickness, level of invasion and node dissection in stage I cutaneous melanoma. Ann Surg 182:572–575PubMed Breslow A (1975) Tumor thickness, level of invasion and node dissection in stage I cutaneous melanoma. Ann Surg 182:572–575PubMed
8.
go back to reference Balch CM et al (2001) Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 19:3635–3648PubMed Balch CM et al (2001) Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma. J Clin Oncol 19:3635–3648PubMed
9.
go back to reference Eton O et al (1998) Prognostic factors for survival of patients treated systemically for disseminated melanoma. J Clin Oncol 16:1103–1111PubMed Eton O et al (1998) Prognostic factors for survival of patients treated systemically for disseminated melanoma. J Clin Oncol 16:1103–1111PubMed
10.
go back to reference Francken AB et al (2008) Prognosis and determinants of outcome following locoregional or distant recurrence in patients with cutaneous melanoma. Ann Surg Oncol 15:1476–1484PubMed Francken AB et al (2008) Prognosis and determinants of outcome following locoregional or distant recurrence in patients with cutaneous melanoma. Ann Surg Oncol 15:1476–1484PubMed
11.
go back to reference Thompson JF, Scolyer RA, Kefford RF (2009) Cutaneous melanoma in the era of molecular profiling. Lancet 374:362–365PubMed Thompson JF, Scolyer RA, Kefford RF (2009) Cutaneous melanoma in the era of molecular profiling. Lancet 374:362–365PubMed
12.
go back to reference Singh AD et al (2004) The Zimmerman-McLean-Foster hypothesis: 25 years later. Br J Ophthalmol 88:962–967PubMed Singh AD et al (2004) The Zimmerman-McLean-Foster hypothesis: 25 years later. Br J Ophthalmol 88:962–967PubMed
13.
go back to reference White RM, Zon LI (2008) Melanocytes in development, regeneration, and cancer. Cell Stem Cell 3:242–252PubMed White RM, Zon LI (2008) Melanocytes in development, regeneration, and cancer. Cell Stem Cell 3:242–252PubMed
14.
go back to reference Roberts DL et al (2002) U.K. guidelines for the management of cutaneous melanoma. Br J Dermatol 146:7–17PubMed Roberts DL et al (2002) U.K. guidelines for the management of cutaneous melanoma. Br J Dermatol 146:7–17PubMed
15.
go back to reference Michaelson JS et al (2005) Spread of human cancer cells occurs with probabilities indicative of a nongenetic mechanism. Br J Cancer 93:1244–1249PubMed Michaelson JS et al (2005) Spread of human cancer cells occurs with probabilities indicative of a nongenetic mechanism. Br J Cancer 93:1244–1249PubMed
16.
go back to reference Wascher RA et al (2003) Molecular tumor markers in the blood: early prediction of disease outcome in melanoma patients treated with a melanoma vaccine. J Clin Oncol 21:2558–2563PubMed Wascher RA et al (2003) Molecular tumor markers in the blood: early prediction of disease outcome in melanoma patients treated with a melanoma vaccine. J Clin Oncol 21:2558–2563PubMed
17.
go back to reference Hoek KS et al (2006) Metastatic potential of melanomas defined by specific gene expression profiles with no BRAF signature. Pigment Cell Res 19:290–302PubMed Hoek KS et al (2006) Metastatic potential of melanomas defined by specific gene expression profiles with no BRAF signature. Pigment Cell Res 19:290–302PubMed
18.
go back to reference Bockhorn M, Jain RK, Munn LL (2007) Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol 8:444–448PubMed Bockhorn M, Jain RK, Munn LL (2007) Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol 8:444–448PubMed
19.
go back to reference Chen LL et al (2009) Cancer metastasis networks and the prediction of progression patterns. Br J Cancer 101:749–758PubMed Chen LL et al (2009) Cancer metastasis networks and the prediction of progression patterns. Br J Cancer 101:749–758PubMed
20.
go back to reference Butler TP, Gullino PM (1975) Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Res 35:512–516PubMed Butler TP, Gullino PM (1975) Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Res 35:512–516PubMed
21.
go back to reference Fidler IJ et al (2002) The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol 3:53–57PubMed Fidler IJ et al (2002) The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol 3:53–57PubMed
22.
go back to reference Chiang AC, Massague J (2008) Molecular basis of metastasis. N Engl J Med 359:2814–2823PubMed Chiang AC, Massague J (2008) Molecular basis of metastasis. N Engl J Med 359:2814–2823PubMed
23.
go back to reference Ulmer A et al (2008) Visualization of circulating melanoma cells in peripheral blood of patients with primary uveal melanoma. Clin Cancer Res 14:4469–4474PubMed Ulmer A et al (2008) Visualization of circulating melanoma cells in peripheral blood of patients with primary uveal melanoma. Clin Cancer Res 14:4469–4474PubMed
24.
go back to reference Ulmer A et al (2004) Immunomagnetic enrichment, genomic characterization, and prognostic impact of circulating melanoma cells. Clin Cancer Res 10:531–537PubMed Ulmer A et al (2004) Immunomagnetic enrichment, genomic characterization, and prognostic impact of circulating melanoma cells. Clin Cancer Res 10:531–537PubMed
25.
go back to reference Husemann Y et al (2008) Systemic spread is an early step in breast cancer. Cancer Cell 13:58–68PubMed Husemann Y et al (2008) Systemic spread is an early step in breast cancer. Cancer Cell 13:58–68PubMed
26.
go back to reference Mocellin S et al (2006) The prognostic value of circulating tumor cells in patients with melanoma: a systematic review and meta-analysis. Clin Cancer Res 12:4605–4613PubMed Mocellin S et al (2006) The prognostic value of circulating tumor cells in patients with melanoma: a systematic review and meta-analysis. Clin Cancer Res 12:4605–4613PubMed
27.
go back to reference Sekine I et al (2002) Relationship between objective responses in phase I trials and potential efficacy of non-specific cytotoxic investigational new drugs. Ann Oncol 13:1300–1306PubMed Sekine I et al (2002) Relationship between objective responses in phase I trials and potential efficacy of non-specific cytotoxic investigational new drugs. Ann Oncol 13:1300–1306PubMed
28.
go back to reference Bhatia S, Tykodi SS, Thompson JA (2009) Treatment of metastatic melanoma: an overview. Oncol 23:488–496 Bhatia S, Tykodi SS, Thompson JA (2009) Treatment of metastatic melanoma: an overview. Oncol 23:488–496
29.
go back to reference T FR Crosby, Coles B, Mason M (2000) Systemic treatments for metastatic cutaneous melanoma. Cochrane Database Syst Rev 2:CD001215 T FR Crosby, Coles B, Mason M (2000) Systemic treatments for metastatic cutaneous melanoma. Cochrane Database Syst Rev 2:CD001215
30.
go back to reference Richards JM BA, Gonzalez R, et al. (2005) High-dose Allovectin-7® in patients with advanced metastatic melanoma: final Phase 2 data and design of Phase 3 registration trial. J Clin Oncol 23:16s:7543 Richards JM BA, Gonzalez R, et al. (2005) High-dose Allovectin-7® in patients with advanced metastatic melanoma: final Phase 2 data and design of Phase 3 registration trial. J Clin Oncol 23:16s:7543
31.
go back to reference Shepherd C, Puzanov I, Sosman JA (2010) B-RAF inhibitors: an evolving role in the therapy of malignant melanoma. Curr Oncol Rep 12:146–152PubMed Shepherd C, Puzanov I, Sosman JA (2010) B-RAF inhibitors: an evolving role in the therapy of malignant melanoma. Curr Oncol Rep 12:146–152PubMed
32.
go back to reference Fisher DE et al (2010) Melanoma from bench to bedside: meeting report from the 6th international melanoma congress. Pigment Cell Melanoma Res 23:14–26PubMed Fisher DE et al (2010) Melanoma from bench to bedside: meeting report from the 6th international melanoma congress. Pigment Cell Melanoma Res 23:14–26PubMed
33.
go back to reference Modjtahedi H, Essapen S (2009) Epidermal growth factor receptor inhibitors in cancer treatment: advances, challenges and opportunities. Anticancer Drugs 20:851–865PubMed Modjtahedi H, Essapen S (2009) Epidermal growth factor receptor inhibitors in cancer treatment: advances, challenges and opportunities. Anticancer Drugs 20:851–865PubMed
34.
go back to reference Flaherty KT et al (2010) Inhibition of Mutated, Activated BRAF in Metastatic Melanoma. N Engl J Med 363:809–819PubMed Flaherty KT et al (2010) Inhibition of Mutated, Activated BRAF in Metastatic Melanoma. N Engl J Med 363:809–819PubMed
35.
go back to reference Mazzocca A, Carloni V (2009) The metastatic process: methodological advances and pharmacological challenges. Curr Med Chem 16:1704–1717PubMed Mazzocca A, Carloni V (2009) The metastatic process: methodological advances and pharmacological challenges. Curr Med Chem 16:1704–1717PubMed
36.
go back to reference Van Den Bossche K, Naeyaert JM, Lambert J (2006) The quest for the mechanism of melanin transfer. Traffic 7:769–778 Van Den Bossche K, Naeyaert JM, Lambert J (2006) The quest for the mechanism of melanin transfer. Traffic 7:769–778
37.
go back to reference Moustakas A, Heldin CH (2007) Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci 98:1512–1520PubMed Moustakas A, Heldin CH (2007) Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci 98:1512–1520PubMed
38.
go back to reference Paterlini-Brechot P, Benali NL (2007) Circulating tumor cells (CTC) detection: clinical impact and future directions. Cancer Lett 253:180–204PubMed Paterlini-Brechot P, Benali NL (2007) Circulating tumor cells (CTC) detection: clinical impact and future directions. Cancer Lett 253:180–204PubMed
39.
go back to reference Pouyssegur J, Dayan F, Mazure NM (2006) Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441:437–443PubMed Pouyssegur J, Dayan F, Mazure NM (2006) Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature 441:437–443PubMed
40.
go back to reference Kageshita T et al (2001) Loss of beta-catenin expression associated with disease progression in malignant melanoma. Br J Dermatol 145:210–216PubMed Kageshita T et al (2001) Loss of beta-catenin expression associated with disease progression in malignant melanoma. Br J Dermatol 145:210–216PubMed
41.
go back to reference Vogelmann R et al (2005) TGFbeta-induced downregulation of E-cadherin-based cell-cell adhesion depends on PI3-kinase and PTEN. J Cell Sci 118:4901–4912PubMed Vogelmann R et al (2005) TGFbeta-induced downregulation of E-cadherin-based cell-cell adhesion depends on PI3-kinase and PTEN. J Cell Sci 118:4901–4912PubMed
42.
go back to reference Peinado H, Portillo F, Cano A (2004) Transcriptional regulation of cadherins during development and carcinogenesis. Int J Dev Biol 48:365–375PubMed Peinado H, Portillo F, Cano A (2004) Transcriptional regulation of cadherins during development and carcinogenesis. Int J Dev Biol 48:365–375PubMed
43.
go back to reference Hsu MY et al (2000) E-cadherin expression in melanoma cells restores keratinocyte-mediated growth control and down-regulates expression of invasion-related adhesion receptors. Am J Pathol 156:1515–1525PubMed Hsu MY et al (2000) E-cadherin expression in melanoma cells restores keratinocyte-mediated growth control and down-regulates expression of invasion-related adhesion receptors. Am J Pathol 156:1515–1525PubMed
44.
go back to reference Sanz-Moreno V et al (2008) Rac activation and inactivation control plasticity of tumor cell movement. Cell 135:510–523PubMed Sanz-Moreno V et al (2008) Rac activation and inactivation control plasticity of tumor cell movement. Cell 135:510–523PubMed
45.
go back to reference Egeblad M, Werb Z (2002) New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2:161–174PubMed Egeblad M, Werb Z (2002) New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2:161–174PubMed
46.
go back to reference Lewis CE, Pollard JW (2006) Distinct role of macrophages in different tumor microenvironments. Cancer Res 66:605–612PubMed Lewis CE, Pollard JW (2006) Distinct role of macrophages in different tumor microenvironments. Cancer Res 66:605–612PubMed
47.
go back to reference Liotta LA, Kleinerman J, Saidel GM (1974) Quantitative relationships of intravascular tumor cells, tumor vessels, and pulmonary metastases following tumor implantation. Cancer Res 34:997–1004PubMed Liotta LA, Kleinerman J, Saidel GM (1974) Quantitative relationships of intravascular tumor cells, tumor vessels, and pulmonary metastases following tumor implantation. Cancer Res 34:997–1004PubMed
48.
go back to reference Glinsky GV (1997) Apoptosis in metastatic cancer cells. Crit Rev Oncol Hematol 25:175–186PubMed Glinsky GV (1997) Apoptosis in metastatic cancer cells. Crit Rev Oncol Hematol 25:175–186PubMed
49.
go back to reference Swartz MA et al (1999) Cells shed from tumours show reduced clonogenicity, resistance to apoptosis, and in vivo tumorigenicity. Br J Cancer 81:756–759PubMed Swartz MA et al (1999) Cells shed from tumours show reduced clonogenicity, resistance to apoptosis, and in vivo tumorigenicity. Br J Cancer 81:756–759PubMed
50.
go back to reference Larson CJ et al (2004) Apoptosis of circulating tumor cells in prostate cancer patients. Cytometry A 62:46–53PubMed Larson CJ et al (2004) Apoptosis of circulating tumor cells in prostate cancer patients. Cytometry A 62:46–53PubMed
51.
go back to reference Mehes G et al (2001) Circulating breast cancer cells are frequently apoptotic. Am J Pathol 159:17–20PubMed Mehes G et al (2001) Circulating breast cancer cells are frequently apoptotic. Am J Pathol 159:17–20PubMed
52.
go back to reference Holmgren L, O’Reilly MS, Folkman J (1995) Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1:149–153PubMed Holmgren L, O’Reilly MS, Folkman J (1995) Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1:149–153PubMed
53.
go back to reference Nguyen DX, Bos PD, Massague J (2009) Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9:274–284PubMed Nguyen DX, Bos PD, Massague J (2009) Metastasis: from dissemination to organ-specific colonization. Nat Rev Cancer 9:274–284PubMed
54.
go back to reference Borsig L et al (2002) Synergistic effects of L- and P-selectin in facilitating tumor metastasis can involve non-mucin ligands and implicate leukocytes as enhancers of metastasis. Proc Natl Acad Sci USA 99:2193–2198PubMed Borsig L et al (2002) Synergistic effects of L- and P-selectin in facilitating tumor metastasis can involve non-mucin ligands and implicate leukocytes as enhancers of metastasis. Proc Natl Acad Sci USA 99:2193–2198PubMed
55.
go back to reference Laubli H et al (2006) L-selectin facilitation of metastasis involves temporal induction of Fut7-dependent ligands at sites of tumor cell arrest. Cancer Res 66:1536–1542PubMed Laubli H et al (2006) L-selectin facilitation of metastasis involves temporal induction of Fut7-dependent ligands at sites of tumor cell arrest. Cancer Res 66:1536–1542PubMed
56.
go back to reference Fuertes MB et al (2008) Intracellular retention of the NKG2D ligand MHC class I chain-related gene A in human melanomas confers immune privilege and prevents NK cell-mediated cytotoxicity. J Immunol 180:4606–4614PubMed Fuertes MB et al (2008) Intracellular retention of the NKG2D ligand MHC class I chain-related gene A in human melanomas confers immune privilege and prevents NK cell-mediated cytotoxicity. J Immunol 180:4606–4614PubMed
57.
go back to reference Lacreusette A et al (2007) Loss of oncostatin M receptor beta in metastatic melanoma cells. Oncogene 26:881–892PubMed Lacreusette A et al (2007) Loss of oncostatin M receptor beta in metastatic melanoma cells. Oncogene 26:881–892PubMed
58.
go back to reference Horak CE et al (2008) The role of metastasis suppressor genes in metastatic dormancy. APMIS 116:586–601PubMed Horak CE et al (2008) The role of metastasis suppressor genes in metastatic dormancy. APMIS 116:586–601PubMed
59.
go back to reference Eyles J et al (2010) Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J Clin Invest 120:2030–2039PubMed Eyles J et al (2010) Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. J Clin Invest 120:2030–2039PubMed
60.
go back to reference Yoshida BA et al (2000) Metastasis-suppressor genes: a review and perspective on an emerging field. J Natl Cancer Inst 92:1717–1730PubMed Yoshida BA et al (2000) Metastasis-suppressor genes: a review and perspective on an emerging field. J Natl Cancer Inst 92:1717–1730PubMed
61.
go back to reference Röcken M (2010) Early tumor dissemination, but late metastasis: insights into tumor dormancy. J Clin Invest 120:1800–1803PubMed Röcken M (2010) Early tumor dissemination, but late metastasis: insights into tumor dormancy. J Clin Invest 120:1800–1803PubMed
62.
go back to reference Harms JF, Welch DR, Miele ME (2003) KISS1 metastasis suppression and emergent pathways. Clin Exp Metastasis 20:11–18PubMed Harms JF, Welch DR, Miele ME (2003) KISS1 metastasis suppression and emergent pathways. Clin Exp Metastasis 20:11–18PubMed
63.
go back to reference Boissan M, Poupon MF, Lacombe ML (2007) NM23 and metastasis suppressor genes: update. Med Sci (Paris) 23:1115–1123 Boissan M, Poupon MF, Lacombe ML (2007) NM23 and metastasis suppressor genes: update. Med Sci (Paris) 23:1115–1123
64.
go back to reference Steeg PS, Horak CE, Miller KD (2008) Clinical-translational approaches to the Nm23–H1 metastasis suppressor. Clin Cancer Res 14:5006–5012PubMed Steeg PS, Horak CE, Miller KD (2008) Clinical-translational approaches to the Nm23–H1 metastasis suppressor. Clin Cancer Res 14:5006–5012PubMed
65.
go back to reference Gobeil S et al (2008) A genome-wide shRNA screen identifies GAS1 as a novel melanoma metastasis suppressor gene. Genes Dev 22:2932–2940PubMed Gobeil S et al (2008) A genome-wide shRNA screen identifies GAS1 as a novel melanoma metastasis suppressor gene. Genes Dev 22:2932–2940PubMed
66.
go back to reference Shevde LA et al (2002) Suppression of human melanoma metastasis by the metastasis suppressor gene, BRMS1. Exp Cell Res 273:229–239PubMed Shevde LA et al (2002) Suppression of human melanoma metastasis by the metastasis suppressor gene, BRMS1. Exp Cell Res 273:229–239PubMed
67.
go back to reference Lee CC et al (2008) Improved survival after lymphadenectomy for nodal metastasis from an unknown primary melanoma. J Clin Oncol 26:535–541PubMed Lee CC et al (2008) Improved survival after lymphadenectomy for nodal metastasis from an unknown primary melanoma. J Clin Oncol 26:535–541PubMed
68.
go back to reference Vijuk G, Coates AS (1998) Survival of patients with visceral metastatic melanoma from an occult primary lesion: a retrospective matched cohort study. Ann Oncol 9:419–422PubMed Vijuk G, Coates AS (1998) Survival of patients with visceral metastatic melanoma from an occult primary lesion: a retrospective matched cohort study. Ann Oncol 9:419–422PubMed
69.
go back to reference Zalaudek I et al (2003) Local recurrence in melanoma in situ: influence of sex, age, site of involvement and therapeutic modalities. Br J Dermatol 148:703–708PubMed Zalaudek I et al (2003) Local recurrence in melanoma in situ: influence of sex, age, site of involvement and therapeutic modalities. Br J Dermatol 148:703–708PubMed
70.
go back to reference Koyanagi K et al (2005) Serial monitoring of circulating melanoma cells during neoadjuvant biochemotherapy for stage III melanoma: outcome prediction in a multicenter trial. J Clin Oncol 23:8057–8064PubMed Koyanagi K et al (2005) Serial monitoring of circulating melanoma cells during neoadjuvant biochemotherapy for stage III melanoma: outcome prediction in a multicenter trial. J Clin Oncol 23:8057–8064PubMed
71.
go back to reference Kujala E, Mäkitie T, Kivelä T (2003) Very long-term prognosis of patients with malignant uveal melanoma. Investig Ophthalmol Vis Sci 44:4651–4659 Kujala E, Mäkitie T, Kivelä T (2003) Very long-term prognosis of patients with malignant uveal melanoma. Investig Ophthalmol Vis Sci 44:4651–4659
72.
go back to reference Wargo JA, Tanabe K (2009) Surgical management of melanoma. Hematol Oncol Clin North Am 23:565–581PubMed Wargo JA, Tanabe K (2009) Surgical management of melanoma. Hematol Oncol Clin North Am 23:565–581PubMed
73.
go back to reference Ramaswamy S et al (2003) A molecular signature of metastasis in primary solid tumors. Nat Genet 33:49–54PubMed Ramaswamy S et al (2003) A molecular signature of metastasis in primary solid tumors. Nat Genet 33:49–54PubMed
74.
go back to reference Bernards R, Weinberg RA (2002) A progression puzzle. Nature 418:823PubMed Bernards R, Weinberg RA (2002) A progression puzzle. Nature 418:823PubMed
75.
go back to reference Waghorne C et al (1988) Genetic evidence for progressive selection and overgrowth of primary tumors by metastatic cell subpopulations. Cancer Res 48:6109–6114PubMed Waghorne C et al (1988) Genetic evidence for progressive selection and overgrowth of primary tumors by metastatic cell subpopulations. Cancer Res 48:6109–6114PubMed
76.
go back to reference Fang D et al (2005) A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res 65:9328–9337PubMed Fang D et al (2005) A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res 65:9328–9337PubMed
77.
go back to reference Grichnik JM et al (2006) Melanoma, a tumor based on a mutant stem cell? J Invest Dermatol 126:142–153PubMed Grichnik JM et al (2006) Melanoma, a tumor based on a mutant stem cell? J Invest Dermatol 126:142–153PubMed
78.
go back to reference Monzani E et al (2007) Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur J Cancer 43:935–946PubMed Monzani E et al (2007) Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur J Cancer 43:935–946PubMed
79.
go back to reference Schatton T et al (2008) Identification of cells initiating human melanomas. Nature 451:345–349PubMed Schatton T et al (2008) Identification of cells initiating human melanomas. Nature 451:345–349PubMed
80.
go back to reference Zabierowski SE, Herlyn M (2008) Melanoma stem cells: the dark seed of melanoma. J Clin Oncol 26:2890–2894PubMed Zabierowski SE, Herlyn M (2008) Melanoma stem cells: the dark seed of melanoma. J Clin Oncol 26:2890–2894PubMed
81.
go back to reference Wicha MS (2006) Cancer stem cells and metastasis: lethal seeds. Clin Cancer Res 12:5606–5607PubMed Wicha MS (2006) Cancer stem cells and metastasis: lethal seeds. Clin Cancer Res 12:5606–5607PubMed
82.
go back to reference Pantel K, Alix-Panabieres C, Riethdorf S (2009) Cancer micrometastases. Nat Rev Clin Oncol 6:339–351PubMed Pantel K, Alix-Panabieres C, Riethdorf S (2009) Cancer micrometastases. Nat Rev Clin Oncol 6:339–351PubMed
83.
go back to reference Keshet GI et al (2008) MDR1 expression identifies human melanoma stem cells. Biochem Biophys Res Commun 368:930–936PubMed Keshet GI et al (2008) MDR1 expression identifies human melanoma stem cells. Biochem Biophys Res Commun 368:930–936PubMed
84.
go back to reference Schatton T, Frank MH (2008) Cancer stem cells and human malignant melanoma. Pigment Cell Melanoma Res 21:39–55PubMed Schatton T, Frank MH (2008) Cancer stem cells and human malignant melanoma. Pigment Cell Melanoma Res 21:39–55PubMed
85.
go back to reference Smalley KS, Herlyn M (2009) Integrating tumor-initiating cells into the paradigm for melanoma targeted therapy. Int J Cancer 124:1245–1250PubMed Smalley KS, Herlyn M (2009) Integrating tumor-initiating cells into the paradigm for melanoma targeted therapy. Int J Cancer 124:1245–1250PubMed
86.
go back to reference Hess AR, Seftor EA, Gruman LM, Kinch MS, Seftor RE, Hendrix MJ (2006) VE-cadherin regulates EphA2 in aggressive melanoma cells through a novel signaling pathway: implications for vasculogenic mimicry. Cancer Biol Ther 5:228–233PubMed Hess AR, Seftor EA, Gruman LM, Kinch MS, Seftor RE, Hendrix MJ (2006) VE-cadherin regulates EphA2 in aggressive melanoma cells through a novel signaling pathway: implications for vasculogenic mimicry. Cancer Biol Ther 5:228–233PubMed
87.
go back to reference La Porta C (2009) Cancer stem cells: lessons from melanoma. Stem Cell Rev 5:61–65PubMed La Porta C (2009) Cancer stem cells: lessons from melanoma. Stem Cell Rev 5:61–65PubMed
88.
go back to reference Roesch A et al (2010) A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell 141:583–594PubMed Roesch A et al (2010) A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell 141:583–594PubMed
89.
go back to reference Hoek KS, Goding CR (2010) Cancer stem cells versus phenotype-switching in melanoma. Pigment Cell Melanoma Res 23:746–759PubMed Hoek KS, Goding CR (2010) Cancer stem cells versus phenotype-switching in melanoma. Pigment Cell Melanoma Res 23:746–759PubMed
90.
go back to reference Bennett DC (2008) How to make a melanoma: what do we know of the primary clonal events? Pigment Cell Melanoma Res 21:27–38PubMed Bennett DC (2008) How to make a melanoma: what do we know of the primary clonal events? Pigment Cell Melanoma Res 21:27–38PubMed
91.
go back to reference Bosserhoff AK (2006) Novel biomarkers in malignant melanoma. Clin Chim Acta 367:28–35PubMed Bosserhoff AK (2006) Novel biomarkers in malignant melanoma. Clin Chim Acta 367:28–35PubMed
92.
go back to reference Gogas H et al (2009) Biomarkers in melanoma. Ann Oncol 20(Suppl 6):vi8–vi13PubMed Gogas H et al (2009) Biomarkers in melanoma. Ann Oncol 20(Suppl 6):vi8–vi13PubMed
93.
go back to reference Medic S et al (2007) Molecular markers of circulating melanoma cells. Pigment Cell Res 20:80–91PubMed Medic S et al (2007) Molecular markers of circulating melanoma cells. Pigment Cell Res 20:80–91PubMed
94.
go back to reference Koyanagi K et al (2006) Association of circulating tumor cells with serum tumor-related methylated DNA in peripheral blood of melanoma patients. Cancer Res 66:6111–6117PubMed Koyanagi K et al (2006) Association of circulating tumor cells with serum tumor-related methylated DNA in peripheral blood of melanoma patients. Cancer Res 66:6111–6117PubMed
95.
go back to reference Xi L et al (2007) Optimal markers for real-time quantitative reverse transcription PCR detection of circulating tumor cells from melanoma, breast, colon, esophageal, head and neck, and lung cancers. Clin Chem 53:1206–1215PubMed Xi L et al (2007) Optimal markers for real-time quantitative reverse transcription PCR detection of circulating tumor cells from melanoma, breast, colon, esophageal, head and neck, and lung cancers. Clin Chem 53:1206–1215PubMed
96.
go back to reference Koyanagi K et al (2006) Microphthalmia transcription factor as a molecular marker for circulating tumor cell detection in blood of melanoma patients. Clin Cancer Res 12:1137–1143PubMed Koyanagi K et al (2006) Microphthalmia transcription factor as a molecular marker for circulating tumor cell detection in blood of melanoma patients. Clin Cancer Res 12:1137–1143PubMed
97.
go back to reference Alonso SR et al (2007) A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. Cancer Res 67:3450–3460PubMed Alonso SR et al (2007) A high-throughput study in melanoma identifies epithelial-mesenchymal transition as a major determinant of metastasis. Cancer Res 67:3450–3460PubMed
98.
go back to reference Mandruzzato S et al (2006) A gene expression signature associated with survival in metastatic melanoma. J Transl Med 4:50PubMed Mandruzzato S et al (2006) A gene expression signature associated with survival in metastatic melanoma. J Transl Med 4:50PubMed
99.
go back to reference Talantov D et al (2005) Novel genes associated with malignant melanoma but not benign melanocytic lesions. Clin Cancer Res 11:7234–7242PubMed Talantov D et al (2005) Novel genes associated with malignant melanoma but not benign melanocytic lesions. Clin Cancer Res 11:7234–7242PubMed
100.
go back to reference Tucci MG et al (2007) Involvement of E-cadherin, beta-catenin, Cdc42 and CXCR4 in the progression and prognosis of cutaneous melanoma. Br J Dermatol 157:1212–1216PubMed Tucci MG et al (2007) Involvement of E-cadherin, beta-catenin, Cdc42 and CXCR4 in the progression and prognosis of cutaneous melanoma. Br J Dermatol 157:1212–1216PubMed
101.
go back to reference Kauffmann A et al (2008) High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene 27:565–573PubMed Kauffmann A et al (2008) High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene 27:565–573PubMed
102.
go back to reference Winnepenninckx V et al (2006) Gene expression profiling of primary cutaneous melanoma and clinical outcome. J Natl Cancer Inst 98:472–482PubMed Winnepenninckx V et al (2006) Gene expression profiling of primary cutaneous melanoma and clinical outcome. J Natl Cancer Inst 98:472–482PubMed
103.
go back to reference Eberle J et al (2008) Apoptosis pathways and oncolytic adenoviral vectors: promising targets and tools to overcome therapy resistance of malignant melanoma. Exp Dermatol 17:1–11PubMed Eberle J et al (2008) Apoptosis pathways and oncolytic adenoviral vectors: promising targets and tools to overcome therapy resistance of malignant melanoma. Exp Dermatol 17:1–11PubMed
104.
go back to reference Medic S, Ziman M (2009) PAX3 across the spectrum: from melanoblast to melanoma. Crit Rev Biochem Mol Biol 44:85–97PubMed Medic S, Ziman M (2009) PAX3 across the spectrum: from melanoblast to melanoma. Crit Rev Biochem Mol Biol 44:85–97PubMed
105.
go back to reference Adams JM, Strasser A (2008) Is tumor growth sustained by rare cancer stem cells or dominant clones? Cancer Res 68:4018–4021PubMed Adams JM, Strasser A (2008) Is tumor growth sustained by rare cancer stem cells or dominant clones? Cancer Res 68:4018–4021PubMed
106.
go back to reference Carreira S et al (2006) Mitf regulation of Dia1 controls melanoma proliferation and invasiveness. Genes Dev 20:3426–3439PubMed Carreira S et al (2006) Mitf regulation of Dia1 controls melanoma proliferation and invasiveness. Genes Dev 20:3426–3439PubMed
107.
go back to reference Gupta PB et al (2005) The evolving portrait of cancer metastasis. Cold Spring Harb Symp Quant Biol 70:291–297PubMed Gupta PB et al (2005) The evolving portrait of cancer metastasis. Cold Spring Harb Symp Quant Biol 70:291–297PubMed
108.
go back to reference McArdle L et al (2005) Microarray analysis of phosphatase gene expression in human melanoma. Br J Dermatol 152:925–930PubMed McArdle L et al (2005) Microarray analysis of phosphatase gene expression in human melanoma. Br J Dermatol 152:925–930PubMed
109.
go back to reference Topczewska JM et al (2006) Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med 12:925–932PubMed Topczewska JM et al (2006) Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness. Nat Med 12:925–932PubMed
110.
go back to reference Kubic JD et al (2008) Pigmentation PAX-ways: the role of Pax3 in melanogenesis, melanocyte stem cell maintenance, and disease. Pigment Cell Melanoma Res 21:627–645PubMed Kubic JD et al (2008) Pigmentation PAX-ways: the role of Pax3 in melanogenesis, melanocyte stem cell maintenance, and disease. Pigment Cell Melanoma Res 21:627–645PubMed
111.
go back to reference Lang D et al (2005) Pax3 functions at a nodal point in melanocyte stem cell differentiation. Nature 433:884–887PubMed Lang D et al (2005) Pax3 functions at a nodal point in melanocyte stem cell differentiation. Nature 433:884–887PubMed
112.
go back to reference Plummer RS et al (2008) PAX3 expression in primary melanomas and nevi. Mod Pathol 21:525–530PubMed Plummer RS et al (2008) PAX3 expression in primary melanomas and nevi. Mod Pathol 21:525–530PubMed
113.
go back to reference Meng S et al (2004) Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res 10:8152–8162PubMed Meng S et al (2004) Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res 10:8152–8162PubMed
114.
go back to reference Cools-Lartigue JJ et al (2008) Immunomagnetic isolation and in vitro expansion of human uveal melanoma cell lines. Mol Vis 14:50–55PubMed Cools-Lartigue JJ et al (2008) Immunomagnetic isolation and in vitro expansion of human uveal melanoma cell lines. Mol Vis 14:50–55PubMed
115.
go back to reference Quintana E et al (2008) Efficient tumour formation by single human melanoma cells. Nature 456:593–598PubMed Quintana E et al (2008) Efficient tumour formation by single human melanoma cells. Nature 456:593–598PubMed
116.
go back to reference Lewis KD et al (2008) A phase II study of the heparanase inhibitor PI-88 in patients with advanced melanoma. Invest New Drugs 26:89–94PubMed Lewis KD et al (2008) A phase II study of the heparanase inhibitor PI-88 in patients with advanced melanoma. Invest New Drugs 26:89–94PubMed
117.
go back to reference Tewes M et al (2009) Molecular profiling and predictive value of circulating tumor cells in patients with metastatic breast cancer: an option for monitoring response to breast cancer related therapies. Breast Cancer Res Treat 115:581–590PubMed Tewes M et al (2009) Molecular profiling and predictive value of circulating tumor cells in patients with metastatic breast cancer: an option for monitoring response to breast cancer related therapies. Breast Cancer Res Treat 115:581–590PubMed
118.
go back to reference Fehm T et al (2008) Micrometastatic spread in breast cancer: detection, molecular characterization and clinical relevance. Breast Cancer Res 10(Suppl 1):S1PubMed Fehm T et al (2008) Micrometastatic spread in breast cancer: detection, molecular characterization and clinical relevance. Breast Cancer Res 10(Suppl 1):S1PubMed
119.
go back to reference Cristofanilli M et al (2004) Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 351:781–791PubMed Cristofanilli M et al (2004) Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 351:781–791PubMed
120.
go back to reference Dawood S, Cristofanilli M (2007) Integrating circulating tumor cell assays into the management of breast cancer. Curr Treatment Options Oncol 8:89–95 Dawood S, Cristofanilli M (2007) Integrating circulating tumor cell assays into the management of breast cancer. Curr Treatment Options Oncol 8:89–95
Metadata
Title
Genetic factors in metastatic progression of cutaneous melanoma: the future role of circulating melanoma cells in prognosis and management
Authors
A. Ireland
M. Millward
R. Pearce
M. Lee
M. Ziman
Publication date
01-04-2011
Publisher
Springer Netherlands
Published in
Clinical & Experimental Metastasis / Issue 4/2011
Print ISSN: 0262-0898
Electronic ISSN: 1573-7276
DOI
https://doi.org/10.1007/s10585-010-9368-2

Other articles of this Issue 4/2011

Clinical & Experimental Metastasis 4/2011 Go to the issue
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