Skip to main content

OPINION article

Front. Oncol., 12 August 2013
Sec. Cancer Genetics
This article is part of the Research Topic Melanoma genetics/genomics View all 16 articles

Circulating Melanoma Cells: Scoping the Target

  • 1Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
  • 2Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA

Circulating Melanoma Cells

Molecular markers have been increasingly applied in cancer to assess metastatic risk and to guide treatment, including in melanoma, where assessment of BRAF mutations in tumor tissues to determine suitability for treatment with vemurafenib is now routine (1). Static assessment of tumor tissues, however, does not indicate whether tumor cells are being shed or whether treatment is reducing metastasis. Because melanoma metastasizes hematogenously, examination of circulating melanoma cells (CMC) is a logical as well as a convenient alternative to the examination of tumor tissues. A reliable assessment of CMC numbers and molecular signatures could have major clinical impact. The failure to demonstrate a survival advantage for adjuvant treatment might be linked to inadequate disease staging and, consequently, inadequate assessment of relapse risk. Because CMC may indicate systemic subclinical disease, their detection and analysis may be useful not only for staging/prognosis but also for assessing response to adjuvant therapy. The discovery of CMC theoretically would also allow for earlier detection of metastasis. This could potentially increase the effectiveness of existing therapies. Serial CMC assessments during treatment may allow for the earlier assessment of response, sparing non-responding patients toxicities. Serial CMC assessments could help determine the mechanisms of resistance and suggest interventions to address them. Furthermore, not all patients with melanoma are candidates for surgery to obtain tissue for analysis of molecular markers, and CMC would provide a liquid biopsy of sum total of tumors at all the sites in the patient.

Polymerase Chain Reaction Approaches

Polymerase chain reaction (PCR)-based techniques that detect the expression of the mRNA/transcripts of melanocyte-associated factors, such as tyrosinase and Melan-A, in nucleated blood cells and present in cell free fraction have been best studied clinically. They have demonstrated promise in melanoma surveillance and in monitoring adjuvant and metastatic therapy (24). They can also be combined with assessment of circulating DNA for melanoma-associated mutations, which in itself can be used to infer the presence of CMC (5). No PCR-based approach, however, has been validated for clinical use, due to limitations in consistency and high false negative rates. These approaches cannot quantify the number of CMC, and morphologic evaluation of the cells cannot be obtained. The presence of normal cellular transcripts by leukocytes, which contribute most of the total nucleotides extracted, may dilute those that are tumor-related, even following substantial enrichment for CMC (6). Furthermore, tumor heterogeneity may lead to clones of cells that do not express the melanocyte marker. There are technical issues. RNA is inherently labile (7). Differences in the PCR methodologies applied as well as differences in data interpretation may also be responsible for the disparate findings of various studies. Because cells are not captured, the ability to evaluate changes in targets or biological characteristics is limited, particularly in the context of the tumor heterogeneity that characterizes melanoma (8).

Cytometric Approaches

Techniques that isolate and enumerate morphologically identified CMC have also been studied. Although several steps can be involved, molecular characterization of the CMC isolated has been accomplished. Typically, cytometric approaches have two components: a preparative and an analytical one. Preparative enrichment is required because CMC are rare in the blood, at counts lower than 10/ml of whole blood (as low as 1 CMC per 1,000,000 leukocytes). The goal is to increase sensitivity. The analytical step eliminates the non-relevant blood cells in the enriched fraction. The goal is to increase specificity. Here the greatest concern is again false negative results. Despite an abundance of potential markers, a consensus on how melanoma cells circulate, their phenotype, and the optimal capture reagent have not been established. Tumor cells in circulation may not always exhibit the criteria used to identify them in the context of tissue biopsy. The CellSearch® system (Veridex LLC, Raritan, NJ, USA), a cytometric approach based on the immunomagnetic capture of circulating EpCAM-positive tumor cells, has been approved by the U.S. Food and Drug Administration to monitor the effectiveness of therapy in patients with metastatic breast, colorectal, and prostate carcinomas (9). No cytometric method has been validated for clinical use in melanoma. Representative approaches applied include the following:

Physical

Density gradient separations with for example, Ficoll, combined with elimination of erythrocytes using isotonic ammonium chloride lysis method have been applied (10). The advantages of these approaches are simplicity and lower costs. However, these techniques typically have unacceptably high cell losses and thus lack sensitivity. Several platforms, including filter-based micro-devices, or microfluidic devices, using size as the capture method have been described (11, 12). Given their heterogeneity, it is not clear that large size is a sufficient criterion to capture all CMC. Dielectrophoretic forces have been applied, and cells of different types have been separated, without interfering with their viability, according to their dielectric and hydrodynamic flow properties (13). These approaches may be applicable to CMC but have not yet been effectively applied clinically.

Immunomagnetic – Positive Selection

Circulating melanoma cells have been positively selected using immunomagnetic cell enrichment technique with antibody to, for example, the high molecular weight-melanoma-associated antigen (HMW-MAA). Cells isolated have been assessed for BRAF status (14). CellSearch® technology has also been applied to CMC (15). Magnetic particles are tagged with antibodies against the melanoma-associated cell-surface antigen CD146 and captured in a magnet. These cells are detected using microscopy by fluorescently tagged antibody to HMWMAA (15). The caveat of this technology is the non-specificity of the capture antigen. CD146 is also expressed on circulating endothelial cells. It is also not known whether all CMC express CD146. Hence, sensitivity of this approach is unclear. Multiple antibody-bound beads were suggested to increase the sensitivity of positive selection of CMCs, however, the approach may add to the complexity of the microscopic image analysis and interpretation (16).

Immunomagnetic – Negative Selection

Negative immunomagnetic selection is an attractive approach for isolating CMC in the absence of reliable CMC surface markers. Antibodies tagged with magnetic particles against CD45 antigen present ubiquitously on leukocytes are used to magnetically deplete a blood sample of white cells. These remaining non-magnetic cells are analyzed for CMC with melanocyte-associated markers, such as Melan-A/MART-1, HMB-45, and S100B. The advantage of this technique is unbiased capture of non-leukocytic cells, and disadvantage is lower purity of the CMC due to less than 100% capture of leukocytes. Negative separation has been successfully used to isolate CMC, and also offers the possibility of molecular characterization (17, 18). Although CMC prepared by this method are not pure, due to less than complete depletion of leukocytes, this disadvantage may be outweighed by the presumed complete capture of all types of CMC.

Automated Cytometric Methods

Flow cytometry methods using antibodies against melanocyte determinant have been used to identify and capture CMC. However, the throughput is low, and the rarity of CMC make this technique by itself less practical (19). High speed scanning microscopy techniques such as fiber-optic based automated scanning technology and laser microdissection may be used in identifying and enumerating CMC that are identified with fluorescently tagged melanocyte-associated markers (20, 21). Combining these technologies with unique chip-based substrates has made possible the molecular characterization of single melanoma cells (21).

Summary

Polymerase chain reaction-based approaches to enumerate rare and heterogeneous CMC have demonstrated promise but do not allow for morphologic or molecular analysis of specific cell populations. Although cytometric approaches are in clinical use in the management of patients with carcinomas, the development of similarly approved technology for CMC has proven challenging because of the lack of specific, cell-surface, CMC capture antigen(s). Analysis of the captured cells, such as the identification of molecular targets or special biological characteristics, with current methods can also be cumbersome. Thus, there remains a need for the development of a reliable, efficient platform to isolate, enrich, and characterize CMC in blood. Molecular assessments are now impacting melanoma management. Given the multitude of therapeutic targets emerging, whole genome sequencing adapted to enriched CMC obtained from peripheral blood samples will be necessary for meaningful evaluation of therapeutic directions, and given tumor heterogeneity, it will most likely need to be aimed at the single cell level.

Acknowledgments

The authors would like to thank Dr. Ernest C. Borden and Dr. Ronald A. Conlon for helpful suggestions with the manuscript.

References

1. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. BRIM-3 Study Group. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med (2011) 364(26):2507–16. doi:10.1056/NEJMoa1103782

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

2. Reid AL, Millward M, Pearce R, Lee M, Frank MH, Ireland A, et al. Markers of circulating tumour cells in the peripheral blood of patients with melanoma correlate with disease recurrence and progression. Br J Dermatol (2013) 168(1):85–92. doi:10.1111/bjd.12057

CrossRef Full Text

3. Hoshimoto S, Shingai T, Morton DL, Kuo C, Faries MB, Chong K, et al. Association between circulating tumor cells and prognosis in patients with stage III melanoma with sentinel lymph node metastasis in a phase III international multicenter trial. J Clin Oncol (2012) 30(31):3819–26. doi:10.1200/JCO.2011.40.0887

CrossRef Full Text

4. Koyanagi K, O’Day SJ, Boasberg P, Atkins MB, Wang HJ, Gonzalez R, et al. Serial monitoring of circulating tumor cells predicts outcome of induction biochemotherapy plus maintenance biotherapy for metastatic melanoma. Clin Cancer Res (2010) 16(8):2402–8. doi:10.1158/1078-0432.CCR-10-0037

CrossRef Full Text

5. Fusi A, Berdel R, Havemann S, Nonnenmacher A, Keilholz U. Enhanced detection of BRAF-mutants by pre-PCR cleavage of wild-type sequences revealed circulating melanoma cells heterogeneity. Eur J Cancer (2011) 47(13):1971–6. doi:10.1016/j.ejca.2011.04.013

CrossRef Full Text

6. Ring A, Smith IE, Dowsett M. Circulating tumour cells in breast cancer. Lancet Oncol (2004) 5(2):79–88. doi:10.1016/S1470-2045(04)01381-6

CrossRef Full Text

7. Becker S, Becker-Pergola G, Fehm T, Wallwiener D, Solomayer EF. Time is an important factor when processing samples for the detection of disseminated tumor cells in blood/bone marrow by reverse transcription-PCR. Clin Chem (2004) 50(4):785–6. doi:10.1373/clinchem.2003.025510

CrossRef Full Text

8. Wilmott JS, Tembe V, Howle JR, Sharma R, Thompson JF, Rizos H, et al. Intratumoral molecular heterogeneity in a BRAF-mutant, BRAF inhibitor-resistant melanoma: a case illustrating the challenges for personalized medicine. Mol Cancer Ther (2012) 11(12):2704–8. doi:10.1158/1535-7163.MCT-12-0530

CrossRef Full Text

9. Miller MC, Doyle GV, Terstappen LW. Significance of circulating tumor cells detected by the CellSearch system in patients with metastatic breast colorectal and prostate cancer. J Oncol (2010) 2010:617421. doi:10.1155/2010/617421

CrossRef Full Text

10. Schittek B, Blaheta HJ, Flörchinger G, Sauer B, Garbe C. Increased sensitivity for the detection of malignant melanoma cells in peripheral blood using an improved protocol for reverse transcription-polymerase chain reaction. Br J Dermatol (1999) 141(1):37–43. doi:10.1046/j.1365-2133.1999.02918.x

CrossRef Full Text

11. Lin HK, Zheng S, Williams AJ, Balic M, Groshen S, Scher HI, et al. Portable filter-based microdevice for detection and characterization of circulating tumor cells. Clin Cancer Res (2010) 16(20):5011–8. doi:10.1158/1078-0432.CCR-10-1105

CrossRef Full Text

12. Hou HW, Warkiani ME, Khoo BL, Li ZR, Soo RA, Tan DS, et al. Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci Rep (2013) 3:1259. doi:10.1038/srep01259

CrossRef Full Text

13. Becker FF, Wang XB, Huang Y, Pethig R, Vykoukal J, Gascoyne PR. Separation of human breast cancer cells from blood by differential dielectric affinity. Proc Natl Acad Sci USA (1995) 92(3):860–4. doi:10.1073/pnas.92.3.860

CrossRef Full Text

14. Sakaizawa K, Goto Y, Kiniwa Y, Uchiyama A, Harada K, Shimada S, et al. Mutation analysis of BRAF and KIT in circulating melanoma cells at the single cell level. Br J Cancer (2012) 106(5):939–46. doi:10.1038/bjc.2012.12

CrossRef Full Text

15. Khoja L, Lorigan P, Zhou C, Lancashire M, Booth J, Cummings J, et al. Biomarker utility of circulating tumor cells in metastatic cutaneous melanoma. J Invest Dermatol (2013) 133(6):1582–90. doi:10.1038/jid.2012.468

CrossRef Full Text

16. Freeman JB, Gray ES, Millward M, Pearce R, Ziman M. Evaluation of a multi-marker immunomagnetic enrichment assay for the quantification of circulating melanoma cells. J Transl Med (2012) 10:192. doi:10.1186/1479-5876-10-192

CrossRef Full Text

17. Liu Z, Fusi A, Klopocki E, Schmittel A, Tinhofer I, Nonnenmacher A, et al. Negative enrichment by immunomagnetic nanobeads for unbiased characterization of circulating tumor cells from peripheral blood of cancer patients. J Transl Med (2011) 9:70. doi:10.1186/1479-5876-9-70

CrossRef Full Text

18. Tong X, Yang L, Lang J, Zborowski M, Chalmers J. Application of immunomagnetic cell enrichment in combination with RT-PCR for the detection of rare circulating head and neck tumor cells in human peripheral blood. Cytometry B Clin Cytom (2007) 72(5):310–23.

19. Somlo G, Lau SK, Frankel P, Hsieh HB, Liu X, Yang L, et al. Multiple biomarker expression on circulating tumor cells in comparison to tumor tissues from primary and metastatic sites in patients with locally advanced/inflammatory, and stage IV breast, cancer, using a novel detection technology. Breast Cancer Res Treat (2011) 128(1):155–63. doi:10.1007/s10549-011-1508-0

CrossRef Full Text

20. Clawson GA, Kimchi E, Patrick SD, Xin P, Harouaka R, Zheng S, et al. Circulating tumor cells in melanoma patients. PLoS ONE (2012) 7(7):e41052. doi:10.1371/journal.pone.0041052

CrossRef Full Text

21. Hou S, Zhao L, Shen Q, Yu J, Ng C, Kong X, et al. Polymer nanofiber-embedded microchips for detection, isolation, and molecular analysis of single circulating melanoma cells. Angew Chem Int Ed Engl (2013) 52(12):3379–83. doi:10.1002/anie.201208452

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Citation: Joshi P, Zborowski M and Triozzi PL (2013) Circulating melanoma cells: scoping the target. Front. Oncol. 3:189. doi: 10.3389/fonc.2013.00189

Received: 03 June 2013; Accepted: 07 July 2013;
Published online: 12 August 2013.

Edited by:

Mike Eccles, University of Otago, New Zealand

Reviewed by:

Mel Ziman, Edith Cowan University, Australia

Copyright: © 2013 Joshi, Zborowski and Triozzi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: triozzp@ccf.org

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.