1
|
Pekarek L, Sánchez Cedra A, Jaudenes YDY, Ospino LR, Iglesias Pedrejón B, Bernier L, Roberts Cervantes ED, Sánchez Cendra C, Cassinello J, Trasobares L, Quesada-Cortés A, Sáez MA, Álvarez-Mon M, Ortega MA. Paradigm of biomarkers in metastatic melanoma (Review). Oncol Lett 2025; 29:78. [PMID: 39650232 PMCID: PMC11622106 DOI: 10.3892/ol.2024.14824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/15/2024] [Indexed: 12/11/2024] Open
Abstract
Metastatic melanoma is an aggressive and deadly form of skin cancer, known for its rapid ability to spread to other organs. Melanoma metastasis involves several steps: Local invasion, lymphovascular invasion and proliferation to new sites. This process is facilitated by genetic alterations, interactions with the tumor microenvironment and evasion of the immune system. Despite advances in therapies, the 5-year survival rate remains low at ~22.5%. Notably, current research is focused on identifying patients who may benefit from specific treatments, considering factors such as mutational load and programmed death ligand 1 expression. BRAF inhibitors and immune checkpoint inhibitors have improved survival, although numerous patients do not respond or develop resistance, underscoring the need for novel biomarkers to optimize treatment and monitoring of the disease. In summary, the purpose of the present article is to review the different serological, histological, microRNA and circulating tumor cell biomarkers that have proven useful in the diagnosis, follow-up and prognosis of metastatic melanoma. These biomarkers represent a promising area for research and clinical application, with the aim of offering more precise and personalized treatments.
Collapse
Affiliation(s)
- Leonel Pekarek
- Department of Medicine and Medical Specialties, Biomedical Network Research Center on Liver and Digestive Diseases, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Oncology Service, University Hospital of Guadalajara, 19002 Guadalajara, Spain
- Ramón y Cajal Institute for Health Research, 28034 Madrid, Spain
| | | | | | - Linda Rocío Ospino
- Oncology Service, University Hospital of Guadalajara, 19002 Guadalajara, Spain
| | | | - Loreto Bernier
- Oncology Service, University Hospital of Guadalajara, 19002 Guadalajara, Spain
| | | | | | - Javier Cassinello
- Oncology Service, University Hospital of Guadalajara, 19002 Guadalajara, Spain
| | - Lidia Trasobares
- Department of Medicine and Medical Specialties, Biomedical Network Research Center on Liver and Digestive Diseases, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Dermatology Service, Prince of Asturias University Hospital, 28806 Alcalá de Henares, Spain
| | - Alicia Quesada-Cortés
- Department of Medicine and Medical Specialties, Biomedical Network Research Center on Liver and Digestive Diseases, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Dermatology Service, Prince of Asturias University Hospital, 28806 Alcalá de Henares, Spain
| | - Miguel A. Sáez
- Department of Medicine and Medical Specialties, Biomedical Network Research Center on Liver and Digestive Diseases, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute for Health Research, 28034 Madrid, Spain
- Pathological Anatomy Service, Central University Defence Hospital-UAH Madrid, 28801 Alcalá de Henares, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Biomedical Network Research Center on Liver and Digestive Diseases, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute for Health Research, 28034 Madrid, Spain
- Diseases of the Immune System-Service of Rheumatology, Oncology and Internal Medicine, Biomedical Network Research Center on Liver and Digestive Diseases, Hospital Universitario Príncipe de Asturias, 28806 Alcalá de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialties, Biomedical Network Research Center on Liver and Digestive Diseases, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
- Ramón y Cajal Institute for Health Research, 28034 Madrid, Spain
- Cancer Registry and Pathological Anatomy Service, Prince of Asturias University Hospital, 28806 Alcalá de Henares, Spain
| |
Collapse
|
2
|
Recent Developments of Circulating Tumor Cell Analysis for Monitoring Cutaneous Melanoma Patients. Cancers (Basel) 2022; 14:cancers14040859. [PMID: 35205608 PMCID: PMC8870206 DOI: 10.3390/cancers14040859] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Circulating tumor cells (CTCs) originating from cutaneous melanoma patients have been studied for several decades as surrogates for real-time clinical status and disease outcomes. Here, we will review clinical studies from the last 15 years that assessed CTCs and disease outcomes for melanoma patients. Assessment of multiple molecular melanoma-associated antigen (MAA) markers by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) was the most common assay allowing for the improvement of assay sensitivity, to address tumor heterogeneity, and to predict patient outcomes. Multicenter studies demonstrate the utility of CTC assays reducing the bias observed in single-center trials. Recent development of CTC enrichment platforms has provided reproducible methods. CTC assessment enables both multiple mRNAs and DNAs genomic profiling. CTC provides specific important translational information on tumor progression, prediction of treatment response, and survival outcomes for cutaneous melanoma patients. Abstract Circulating tumor cells (CTCs) have been studied using multiple technical approaches for interrogating various cancers, as they allow for the real-time assessment of tumor progression, disease recurrence, treatment response, and tumor molecular profiling without the need for a tumor tissue biopsy. Here, we will review studies from the last 15 years on the assessment of CTCs in cutaneous melanoma patients in relation to different clinical outcomes. The focus will be on CTC detection in blood samples obtained from cutaneous melanoma patients of different clinical stages and treatments utilizing multiple platforms. Assessment of multiple molecular melanoma-associated antigen (MAA) markers by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) was the most common assay allowing for the improvement of assay sensitivity, tumor heterogeneity, and to predict patient outcomes. Multicenter studies demonstrate the utility of CTC assays reducing the bias observed in single- center trials. The recent development of CTC enrichment platforms has provided reproducible methods. CTC assessment enables both multiple mRNAs and DNAs genomic aberration profiling. CTC provides specific important translational information on tumor progression, prediction of treatment response, and survival outcomes for cutaneous melanoma patients. The molecular studies on melanoma CTCs have provided and may set standards for other solid tumor CTC analyses.
Collapse
|
3
|
Danila DC. Liquid biopsy as a cancer biomarker-potential, and challenges. Cancer Biomark 2022. [DOI: 10.1016/b978-0-12-824302-2.00013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
4
|
Honoré N, Galot R, van Marcke C, Limaye N, Machiels JP. Liquid Biopsy to Detect Minimal Residual Disease: Methodology and Impact. Cancers (Basel) 2021; 13:5364. [PMID: 34771526 PMCID: PMC8582541 DOI: 10.3390/cancers13215364] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
One reason why some patients experience recurrent disease after a curative-intent treatment might be the persistence of residual tumor cells, called minimal residual disease (MRD). MRD cannot be identified by standard radiological exams or clinical evaluation. Tumor-specific alterations found in the blood indirectly diagnose the presence of MRD. Liquid biopsies thus have the potential to detect MRD, allowing, among other things, the detection of circulating tumor DNA (ctDNA), circulating tumor cells (CTC), or tumor-specific microRNA. Although liquid biopsy is increasingly studied, several technical issues still limit its clinical applicability: low sensitivity, poor standardization or reproducibility, and lack of randomized trials demonstrating its clinical benefit. Being able to detect MRD could give clinicians a more comprehensive view of the risk of relapse of their patients and could select patients requiring treatment escalation with the goal of improving cancer survival. In this review, we are discussing the different methodologies used and investigated to detect MRD in solid cancers, their respective potentials and issues, and the clinical impacts that MRD detection will have on the management of cancer patients.
Collapse
Affiliation(s)
- Natasha Honoré
- Institute for Experimental and Clinical Research (IREC, Pôle MIRO), Université Catholique de Louvain (UCLouvain) ,1200 Brussels, Belgium; (R.G.); (C.v.M.)
| | - Rachel Galot
- Institute for Experimental and Clinical Research (IREC, Pôle MIRO), Université Catholique de Louvain (UCLouvain) ,1200 Brussels, Belgium; (R.G.); (C.v.M.)
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Cédric van Marcke
- Institute for Experimental and Clinical Research (IREC, Pôle MIRO), Université Catholique de Louvain (UCLouvain) ,1200 Brussels, Belgium; (R.G.); (C.v.M.)
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Nisha Limaye
- Genetics of Autoimmune Diseases and Cancer, de Duve Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Jean-Pascal Machiels
- Institute for Experimental and Clinical Research (IREC, Pôle MIRO), Université Catholique de Louvain (UCLouvain) ,1200 Brussels, Belgium; (R.G.); (C.v.M.)
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
5
|
Michelakis D, Lasithiotakis K, Messaritakis I, Ioannou C, Perisynakis K, Souglakos I, Stamatiou D, Chlouverakis G, de Bree E, Romanos I, Zoras O. A feasibility study of circulating melanoma cells in the perioperative context of hyperthermic isolated limb perfusion (HILP) in 20 patients. Int J Hyperthermia 2021; 38:70-78. [PMID: 33487077 DOI: 10.1080/02656736.2021.1874062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/02/2021] [Accepted: 01/05/2021] [Indexed: 10/22/2022] Open
Abstract
INTRODUCTION Hyperthermic Ιsolated Limb Perfusion using melphalan and TNFα (TM-HILP) is a regional chemotherapy method for advanced melanoma. PURPOSE To explore the feasibility of the study of Circulating Melanoma Cells (CMCs) in the context of acute physiological changes induced by TM-HILP and their association with oncological outcomes. METHODS The study included 20 patients undergoing TM-HILP for unresectable in-transit melanoma of the limbs, stage III(B/C/D). CMCs in the peripheral blood were analyzed at 5-time points from the preoperative day until day 7 from surgery using the following biomarkers: MITF, Tyrosinase mRNA, Melan-A and S100b, through quantitative RT-PCR. RESULTS No CMCs according to Tyrosinase and Melan-A biomarkers were found in any sample. Friedman test showed significant alterations perioperatively for MITF (p < .001) and S100b (p = .001). Pairwise tests showed a significant increase of MITF levels on postoperative day 7 compared with postoperative day 1, intraoperative and preoperative levels (p < .05). Pairwise tests for S100b showed a significant difference between intraoperative sample and postoperative day 7 (p < .0001). Patients who experienced a complete response to TM-HILP (n = 12) had higher mean levels of MITF and the difference was significant at the time point immediately after the operation (0.29 ± 0.27 vs. 0.06 ± 0.06, p = .014) and on postoperative day 1 (1.48 ± 2.24 vs. 0.41 ± 0.65, p = .046). There was no association of MITF or S100b levels with 4-year disease specific survival. CONCLUSION TM-HILP is associated with increased levels of CMCs, but there was no association of this increase with survival. Patients with complete response to HILP demonstrate higher values of MITF shortly after the operation.
Collapse
Affiliation(s)
| | | | | | - Christos Ioannou
- Department of Vascular Surgery, University Hospital of Heraklion, Crete, Greece
| | - Kostas Perisynakis
- Department of Nuclear Medicine, University Hospital of Heraklion, Crete, Greece
| | - Ioannis Souglakos
- Department of Clinical Oncology, University Hospital of Heraklion, Crete, Greece
| | - Dimitrios Stamatiou
- Department of Surgical Oncology, University Hospital of Heraklion, Crete, Greece
| | - Gregory Chlouverakis
- Biostatistics Laboratory, Department of Social Medicine, School of Medicine, University of Crete, Crete, Greece
| | - Eelco de Bree
- Department of Surgical Oncology, University Hospital of Heraklion, Crete, Greece
| | - Ioannis Romanos
- Department of Surgical Oncology, University Hospital of Heraklion, Crete, Greece
| | - Odysseas Zoras
- Department of Surgical Oncology, University Hospital of Heraklion, Crete, Greece
| |
Collapse
|
6
|
Lucci A, Hall CS, Patel SP, Narendran B, Bauldry JB, Royal RE, Karhade M, Upshaw JR, Wargo JA, Glitza IC, Wong MKK, Amaria RN, Tawbi HA, Diab A, Davies MA, Gershenwald JE, Lee JE, Hwu P, Ross MI. Circulating Tumor Cells and Early Relapse in Node-positive Melanoma. Clin Cancer Res 2020; 26:1886-1895. [PMID: 32015020 DOI: 10.1158/1078-0432.ccr-19-2670] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/09/2019] [Accepted: 01/29/2020] [Indexed: 02/03/2023]
Abstract
PURPOSE There is a need for sensitive, reproducible biomarkers for patients with stage III melanoma to guide clinical decision making. Circulating tumor cells (CTCs) can be detected in patients with melanoma; however, there are limited data regarding their significance in stage III disease. The aim of this study was to determine whether CTCs are associated with early relapse in stage III melanoma. EXPERIMENTAL DESIGN We prospectively assessed CTCs at first presentation in clinic (baseline) for 243 patients with stage III melanoma. CTCs were measured using the CellSearch System. Relapse-free survival (RFS) was compared between patients with one or more baseline CTC versus those with no CTCs. Log-rank test and Cox regression analysis were applied to establish associations of CTCs with RFS. RESULTS At least one baseline CTC was identified in 90 of 243 (37%) patients. Forty-five (19%), 67 (28%), 118 (49%), and 13 (5%) patients were stage IIIA, IIIB, IIIC, or IIID, respectively. CTC detection was not associated with substage, or primary tumor characteristics. Multivariable analysis demonstrated that the detection of ≥1 baseline CTC was significantly associated with decreased 6-month RFS [log-rank, P < 0.0001; HR, 3.62, 95% confidence interval (CI), 1.78-7.36; P < 0.0001] and 54-month RFS (log-rank, P = 0.01; HR, 1.69; 95% CI, 1.13-2.54; P = 0.01). CONCLUSIONS ≥1 CTC was independently associated with melanoma relapse, suggesting that CTC assessment may be useful to identify patients at risk for relapse who could derive benefit from adjuvant therapy.
Collapse
Affiliation(s)
- Anthony Lucci
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Carolyn S Hall
- Department of Breast Surgery-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Boomadevi Narendran
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jessica B Bauldry
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Royal
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mandar Karhade
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joshua R Upshaw
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Isabella C Glitza
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael K K Wong
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rodabe N Amaria
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
7
|
Boyer M, Cayrefourcq L, Dereure O, Meunier L, Becquart O, Alix-Panabières C. Clinical Relevance of Liquid Biopsy in Melanoma and Merkel Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12040960. [PMID: 32295074 PMCID: PMC7226137 DOI: 10.3390/cancers12040960] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/14/2022] Open
Abstract
Melanoma and Merkel cell carcinoma are two aggressive skin malignancies with high disease-related mortality and increasing incidence rates. Currently, invasive tumor tissue biopsy is the gold standard for their diagnosis, and no reliable easily accessible biomarker is available to monitor patients with melanoma or Merkel cell carcinoma during the disease course. In these last years, liquid biopsy has emerged as a candidate approach to overcome this limit and to identify biomarkers for early cancer diagnosis, prognosis, therapeutic response prediction, and patient follow-up. Liquid biopsy is a blood-based non-invasive procedure that allows the sequential analysis of circulating tumor cells, circulating cell-free and tumor DNA, and extracellular vesicles. These innovative biosources show similar features as the primary tumor from where they originated and represent an alternative to invasive solid tumor biopsy. In this review, the biology and technical challenges linked to the detection and analysis of the different circulating candidate biomarkers for melanoma and Merkel cell carcinoma are discussed as well as their clinical relevance.
Collapse
Affiliation(s)
- Magali Boyer
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, 34093 Montpellier, France; (M.B.); (L.C.)
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, 34093 Montpellier, France; (M.B.); (L.C.)
| | - Olivier Dereure
- Department of Dermatology and INSERM 1058 Pathogenesis and Control of Chronic Infections, University of Montpellier, 34090 Montpellier, France;
| | - Laurent Meunier
- Department of Dermatology, University of Montpellier, 34090 Montpellier, France; (L.M.); (O.B.)
| | - Ondine Becquart
- Department of Dermatology, University of Montpellier, 34090 Montpellier, France; (L.M.); (O.B.)
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, 34093 Montpellier, France; (M.B.); (L.C.)
- Correspondence: ; Tel.: +33-4-1175-99-31; Fax: +33-4-1175-99-33
| |
Collapse
|
8
|
Schneegans S, Lück L, Besler K, Bluhm L, Stadler JC, Staub J, Greinert R, Volkmer B, Kubista M, Gebhardt C, Sartori A, Irwin D, Serkkola E, Af Hällström T, Lianidou E, Sprenger-Haussels M, Hussong M, Mohr P, Schneider SW, Shaffer J, Pantel K, Wikman H. Pre-analytical factors affecting the establishment of a single tube assay for multiparameter liquid biopsy detection in melanoma patients. Mol Oncol 2020; 14:1001-1015. [PMID: 32246814 PMCID: PMC7191195 DOI: 10.1002/1878-0261.12669] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/11/2020] [Accepted: 03/12/2020] [Indexed: 12/18/2022] Open
Abstract
The combination of liquid biomarkers from a single blood tube can provide more comprehensive information on tumor development and progression in cancer patients compared to single analysis. Here, we evaluated whether a combined analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and circulating cell-free microRNA (miRNA) in total plasma and extracellular vesicles (EV) from the same blood sample is feasible and how the results are influenced by the choice of different blood tubes. Peripheral blood from 20 stage IV melanoma patients and five healthy donors (HD) was collected in EDTA, Streck, and Transfix tubes. Peripheral blood mononuclear cell fraction was used for CTC analysis, whereas plasma and EV fractions were used for ctDNA mutation and miRNA analysis. Mutations in cell-free circulating DNA were detected in 67% of patients, with no significant difference between the tubes. CTC was detected in only EDTA blood and only in 15% of patients. miRNA NGS (next-generation sequencing) results were highly influenced by the collection tubes and could only be performed from EDTA and Streck tubes due to hemolysis in Transfix tubes. No overlap of significantly differentially expressed miRNA (patients versus HD) could be found between the tubes in total plasma, whereas eight miRNA were commonly differentially regulated in the EV fraction. In summary, high-quality CTCs, ctDNA, and miRNA data from a single blood tube can be obtained. However, the choice of blood collection tubes is a critical pre-analytical variable.
Collapse
Affiliation(s)
- Svenja Schneegans
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Lelia Lück
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Katharina Besler
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Leonie Bluhm
- Centre of Dermatology, Elbe Clinics, Buxtehude, Germany
| | - Julia-Christina Stadler
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Germany.,Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Germany
| | - Janina Staub
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Beate Volkmer
- Centre of Dermatology, Elbe Clinics, Buxtehude, Germany
| | - Mikael Kubista
- TATAA Biocenter AB, Gothenburg, Sweden.,Department of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Vestec, Czech Republic
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Germany
| | | | | | | | | | - Evi Lianidou
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, University of Athens, Greece
| | | | - Melanie Hussong
- QIAGEN Inc/GmbH, Frederick, MD, USA.,QIAGEN Inc/GmbH, Hilden, Germany
| | - Peter Mohr
- Centre of Dermatology, Elbe Clinics, Buxtehude, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Germany
| | - Jonathan Shaffer
- QIAGEN Inc/GmbH, Frederick, MD, USA.,QIAGEN Inc/GmbH, Hilden, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
9
|
Zhong X, Zhang H, Zhu Y, Liang Y, Yuan Z, Li J, Li J, Li X, Jia Y, He T, Zhu J, Sun Y, Jiang W, Zhang H, Wang C, Ke Z. Circulating tumor cells in cancer patients: developments and clinical applications for immunotherapy. Mol Cancer 2020; 19:15. [PMID: 31980023 PMCID: PMC6982393 DOI: 10.1186/s12943-020-1141-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/16/2020] [Indexed: 02/08/2023] Open
Abstract
Cancer metastasis is the leading cause of cancer-related death. Circulating tumor cells (CTCs) are shed into the bloodstream from either primary or metastatic tumors during an intermediate stage of metastasis. In recent years, immunotherapy has also become an important focus of cancer research. Thus, to study the relationship between CTCs and immunotherapy is extremely necessary and valuable to improve the treatment of cancer. In this review, based on the advancements of CTC isolation technologies, we mainly discuss the clinical applications of CTCs in cancer immunotherapy and the related immune mechanisms of CTC formation. In order to fully understand CTC formation, sufficiently and completely understood molecular mechanism based on the different immune cells is critical. This understanding is a promising avenue for the development of effective immunotherapeutic strategies targeting CTCs.
Collapse
Affiliation(s)
- Xiaoming Zhong
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hangtian Zhang
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuqing Liang
- The Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Commonwealth of Pennsylvania, USA
| | - Zhuolin Yuan
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiachen Li
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Li
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Li
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yifan Jia
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tian He
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiangyuan Zhu
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Sun
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wengting Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Zhang
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Cheng Wang
- Division of Nephrology, Department of medicine, The Fifth Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Lin SY, Chang SC, Lam S, Ramos RI, Tran K, Ohe S, Salomon MP, Bhagat AAS, Lim CT, Fischer TD, Foshag LJ, Boley CL, O’Day SJ, Hoon DS. Prospective Molecular Profiling of Circulating Tumor Cells from Patients with Melanoma Receiving Combinatorial Immunotherapy. Clin Chem 2020; 66:169-177. [PMID: 31672856 PMCID: PMC7193771 DOI: 10.1373/clinchem.2019.307140] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Blood molecular profiling of circulating tumor cells (CTCs) can enable monitoring of patients with metastatic melanoma during checkpoint inhibitor immunotherapy (CII) and in combination with targeted therapies. We developed a microfluidics-based CTC platform to explore CTC profiling utility in CII-treated patients with melanoma using a melanoma messenger RNA (mRNA)/DNA biomarker panel. METHODS Blood samples (n = 213) were collected prospectively from 75 American Joint Committee on Cancer-staged III/IV melanoma patients during CII treatment and those enriched for CTCs. CTC profiling was performed using 5 known melanoma mRNA biomarkers and BRAF V600E DNA mutation. CTC biomarker status associations with clinical outcomes were assessed. RESULTS CTCs were detected in 88% of blood samples from patients with melanoma. CTC-derived biomarkers and clinical variables analyzed using classification and regression tree analysis revealed that a combination of lactate dehydrogenase, CTC-mRNA biomarkers, and tumor BRAF-mutation status was indicative of clinical outcomes for patients with stage IV melanoma (n = 52). The panel stratified low-risk and high-risk patients, whereby the latter had poor disease-free (P = 0.03) and overall survival (P = 0.02). Incorporation of a DNA biomarker with mRNA profiling increased overall CTC-detection capability by 57% compared to mRNA profiling only. RNA sequencing of isolated CTCs identified significant catenin beta 1 (CTNNB1) overexpression (P <0.01) compared to nondisease donor blood. CTC-CTNNB1 was associated with progressive disease/stable disease compared to complete-responder patient status (P = 0.02). Serial CTC profiling identified subclinical disease in patients who developed progressive disease during treatment/follow-up. CONCLUSIONS CTC-derived mRNA/DNA biomarkers have utility for monitoring CII, targeted, and combinatorial therapies in metastatic melanoma patients.
Collapse
Affiliation(s)
- Selena Y. Lin
- Department of Translational Molecular Medicine, John Wayne
Cancer Institute, Saint John’s Health Center, PHS, Santa Monica, CA
| | - Shu-Ching Chang
- Medical Data Research Center, Providence Saint Joseph
Health, Portland, OR
| | - Stella Lam
- Department of Translational Molecular Medicine, John Wayne
Cancer Institute, Saint John’s Health Center, PHS, Santa Monica, CA
| | - Romela Irene Ramos
- Department of Translational Molecular Medicine, John Wayne
Cancer Institute, Saint John’s Health Center, PHS, Santa Monica, CA
| | - Kevin Tran
- Department of Translational Molecular Medicine, John Wayne
Cancer Institute, Saint John’s Health Center, PHS, Santa Monica, CA
| | - Shuichi Ohe
- Department of Translational Molecular Medicine, John Wayne
Cancer Institute, Saint John’s Health Center, PHS, Santa Monica, CA
| | - Matthew P. Salomon
- Department of Translational Molecular Medicine, John Wayne
Cancer Institute, Saint John’s Health Center, PHS, Santa Monica, CA
| | - Ali Asgar S. Bhagat
- Department of Biomedical Engineering and Department of
Mechanical Engineering, National University of Singapore, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering and Department of
Mechanical Engineering, National University of Singapore, Singapore
| | - Trevan D. Fischer
- Department of Surgical Oncology, John Wayne Cancer
Institute, PHS, Santa Monica, CA
| | - Leland J. Foshag
- Department of Surgical Oncology, John Wayne Cancer
Institute, PHS, Santa Monica, CA
| | - Christine L. Boley
- Department of Immuno-Oncology and Clinical Research, John
Wayne Cancer Institute, PHS, Santa Monica, CA
| | - Steven J. O’Day
- Department of Immuno-Oncology and Clinical Research, John
Wayne Cancer Institute, PHS, Santa Monica, CA
| | - Dave S.B. Hoon
- Department of Translational Molecular Medicine, John Wayne
Cancer Institute, Saint John’s Health Center, PHS, Santa Monica, CA
| |
Collapse
|
11
|
Rothé F, Maetens M, Rouas G, Paesmans M, Van den Eynde M, Van Laethem JL, Vergauwe P, Deboever G, Bareche Y, Vandeputte C, Ignatiadis M, Hendlisz A. CTCs as a prognostic and predictive biomarker for stage II/III Colon Cancer: a companion study to the PePiTA trial. BMC Cancer 2019; 19:304. [PMID: 30943928 PMCID: PMC6446374 DOI: 10.1186/s12885-019-5528-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Adjuvant therapy improves the prognosis of stage II & III colon cancer patients. Unfortunately, most patients do not benefit from this treatment. PePITA (NCT00994864) is a prospective, multicenter, non-randomized study whose primary objective is to predict the outcome of adjuvant therapy in colon cancer. METHODS The primary objective was to determine the prognostic and predictive value of circulating tumor cell (CTC) detection before therapy and after one course of preoperative FOLFOX. RESULTS Out of the 58 first patients accrued in PePiTA trial, 36 patients participated in the CTC companion study, of whom 32 had at least one evaluable sample. Only 5 patients (14, 95% CI = 5-30%) had ≥1 CTC/22.5 ml blood in at least one of the two timepoints with 2 patients having ≥1 CTC/22.5 ml at baseline (6, 95% CI: 1-19%). The detection rate of patients with CTCs at baseline being lower than expected, the inclusion of patients in the PePiTA CTC substudy was stopped. The limited sample size did not allow us to investigate the prognostic and predictive value of CTCs in locally advanced colon cancer. CONCLUSIONS Our data illustrate the need for further standardized studies in order to find the most reliable prognostic/predictive biomarker in early-stage colon cancer. TRIAL REGISTRATION This trial was prospectively registered at Jules Bordet institute ( NCT00994864 ) on the October 14, 2009.
Collapse
Affiliation(s)
- Françoise Rothé
- J.-C. Heuson Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| | - Marion Maetens
- J.-C. Heuson Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Ghizlane Rouas
- J.-C. Heuson Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Marianne Paesmans
- Data centre, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Marc Van den Eynde
- Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | - Philippe Vergauwe
- Department of Gastroenterology, General Hospital Groeninge, Kortrijk, Belgium
| | - Guido Deboever
- Department of Gastroenterology, Digestive Oncology, AZ Damiaan Ziekenhuis, Oostende, Belgium
| | - Yacine Bareche
- J.-C. Heuson Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Caroline Vandeputte
- Gastrointestinal Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Michail Ignatiadis
- Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Alain Hendlisz
- Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
12
|
Seenivasan R, Warrick JW, Rodriguez CI, Mattison W, Beebe DJ, Setaluri V, Gunasekaran S. Integrating Electrochemical Immunosensing and Cell Adhesion Technologies for Cancer Cell Detection and Enumeration. Electrochim Acta 2018; 286:205-211. [PMID: 31130739 PMCID: PMC6530932 DOI: 10.1016/j.electacta.2018.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We have successfully integrated techniques for controlling cell adhesion and performing electrochemical differential pulse voltammetry (DPV) through the use of digitally controlled microfluidics and patterned transparent indium tin oxide electrode arrays to enable rapid and sensitive enumeration of cancer cells in a scalable microscale format. This integrated approach leverages a dual-working electrode (WE) surface to improve the specificity of the detection system. Here, one of the WE surfaces is functionalized with anti-Melanocortin 1 Receptor antibodies specific to melanoma cancer cells, while the other WE acts as a control (i.e., without antibody), for detecting non-specific interactions between cells and the electrode. The method is described and shown to provide effective detection of melanoma cells at concentrations ranging between 25 to 300 cells per 20 μL sample volume after a 5 min incubation and 15 s of DPV measurements. The estimated limit of detection was ~17 cells. The sensitivity and specificity of the assay were quantified using addition of large fractions of non-target cells and resulted in a detection reproducibility of ~97%. The proposed approach demonstrates a unique integration of electrochemical sensing and microfluidic cell adhesion technologies with multiple advantages such as label-free detection, short detection times, and low sample volumes. Next steps for this platform include testing with patient samples and use of other cell-surface biomarkers for detection and enumeration of circulating tumor cells in prostate, breast, and colon cancer.
Collapse
Affiliation(s)
- Rajesh Seenivasan
- Department of Biological Systems Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jay W. Warrick
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Carlos I. Rodriguez
- Department of Dermatology, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - William Mattison
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J. Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Vijayasaradhi Setaluri
- Department of Dermatology, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sundaram Gunasekaran
- Department of Biological Systems Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
13
|
Chakraborty S. Electrokinetics with blood. Electrophoresis 2018; 40:180-189. [DOI: 10.1002/elps.201800353] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/13/2018] [Accepted: 09/14/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Suman Chakraborty
- Department of Mechanical Engineering; Indian Institute of Technology Kharagpur; Kharagpur India
| |
Collapse
|
14
|
Novel frontiers in detecting cancer metastasis. Clin Exp Metastasis 2018; 35:403-412. [PMID: 30022365 DOI: 10.1007/s10585-018-9918-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/21/2018] [Indexed: 12/15/2022]
Abstract
Cancer microenvironment is the critical battle ground between the cancer cells and host response. Thus, more emphasis is directed to study the relationship between cancer cells and the stromal cells. Multiplex microscopy is an emerging technique in which multiple cell populations within the cancer microenvironment may be stained so that spatial relationship between cancer cells and, in particular, the immune cells may be studied during different stages of cancer development. Recent discovery of mutational burden and neoantigens in cancer has opened new landscapes in the interaction of host immune cells and cancer neoantigens. The emerging role of miRNAs may become an added dimension to study cancer beyond traditional pathway of DNA directed RNA being associated with the malignant behavior of cancer. Circulating tumor cells, cancer markers and ctDNA can be used as markers for circulating cancer cells in the blood. Further studies are needed to validate if liquid biopsy of cancer may become a routine clinical tool to screen cancer or follow patients for recurrence or responses to treatment.
Collapse
|
15
|
Hall CS, Ross M, Bowman Bauldry JB, Upshaw J, Karhade MG, Royal R, Patel S, Lucci A. Circulating Tumor Cells in Stage IV Melanoma Patients. J Am Coll Surg 2018; 227:116-124. [PMID: 29746918 DOI: 10.1016/j.jamcollsurg.2018.04.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/16/2018] [Accepted: 04/16/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Management of stage IV melanoma patients remains a challenge. In spite of promising new therapies, many patients develop resistance and progression. The aim of this pilot study was to determine if circulating tumor cells (CTCs) are associated with shortened (180-day) progression-free survival (PFS) after a baseline CTC assessment in stage IV melanoma patients. STUDY DESIGN A baseline CTC assessment was performed in 93 stage IV melanoma patients using a commercially available immunomagnetic system. The presence of 1 or more CTC was considered a positive result. A Cox multivariable regression model was used to evaluate the association between presence of CTCs at baseline and PFS, after adjusting for covariables. Kaplan-Meier curves and a log-rank test were used to summarize and compare unadjusted PFS for patients stratified by CTC positivity. RESULTS Median follow-up was 17 months; mean age was 55 years. Thirteen of 93 (14%) patients had no evidence of disease (NED) at baseline CTC assessment. One or more CTC was detected in 39 of 93 (42%) of patients at baseline; CTCs were not associated with primary melanoma features or NED status. Twenty-eight of 93 (30%) patients progressed within 180 days of baseline draw, with 20 of 39 (51%) of the CTC-positive patients relapsing compared with 8 of 54 (15%) of the CTC-negative patients. In adjusted Cox models, a significant association was found suggesting worse PFS within 180 days for CTC-positive patients at baseline (vs CTC-negative) (hazard ratio 4.69, 95% CI 1.59 to 13.77, p = 0.005). CONCLUSIONS One or more CTCs at baseline were associated with progression within 180 days in stage IV melanoma patients. This information warrants further study of CTCs as a means of identifying patients at high-risk for disease progression.
Collapse
Affiliation(s)
- Carolyn S Hall
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Merrick Ross
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jessica B Bowman Bauldry
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Joshua Upshaw
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mandar G Karhade
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard Royal
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sapna Patel
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anthony Lucci
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
16
|
Yang B, Chen B, He M, Yin X, Xu C, Hu B. Aptamer-Based Dual-Functional Probe for Rapid and Specific Counting and Imaging of MCF-7 Cells. Anal Chem 2018; 90:2355-2361. [PMID: 29308635 DOI: 10.1021/acs.analchem.7b04927] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Development of multimodal detection technologies for accurate diagnosis of cancer at early stages is in great demand. In this work, we report a novel approach using an aptamer-based dual-functional probe for rapid, sensitive, and specific counting and visualization of MCF-7 cells by inductively coupled plasma-mass spectrometry (ICP-MS) and fluorescence imaging. The probe consists of a recognition unit of aptamer to catch cancer cells specifically, a fluorescent dye (FAM) moiety for fluorescence resonance energy transfer (FRET)-based "off-on" fluorescence imaging as well as gold nanoparticles (Au NPs) tag for both ICP-MS quantification and fluorescence quenching. Due to the signal amplification effect and low spectral interference of Au NPs in ICP-MS, an excellent linearity and sensitivity were achieved. Accordingly, a limit of detection of 81 MCF-7 cells and a relative standard deviation of 5.6% (800 cells, n = 7) were obtained. The dynamic linear range was 2 × 102 to 1.2 × 104 cells, and the recoveries in human whole blood were in the range of 98-110%. Overall, the established method provides quantitative and visualized information on MCF-7 cells with a simple and rapid process and paves the way for a promising strategy for biomedical research and clinical diagnostics.
Collapse
Affiliation(s)
- Bin Yang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Beibei Chen
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Man He
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Xiao Yin
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Chi Xu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), Department of Chemistry, Wuhan University , Wuhan 430072, China
| | - Bin Hu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), Department of Chemistry, Wuhan University , Wuhan 430072, China
| |
Collapse
|
17
|
Lin SY, Orozco JIJ, Hoon DSB. Detection of Minimal Residual Disease and Its Clinical Applications in Melanoma and Breast Cancer Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1100:83-95. [PMID: 30411261 DOI: 10.1007/978-3-319-97746-1_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Melanoma and breast cancer (BC) patients face a high risk of recurrence and disease progression after curative surgery and/or therapeutic treatment. Monitoring for minimal residual disease (MRD) during a disease-free follow-up period would greatly improve patient outcomes through earlier detection of relapse or treatment resistance. However, MRD monitoring in solid tumors such as melanoma and BC are not well established. Here, we discuss the clinical applications of MRD monitoring in melanoma and BC patients and highlight the current approaches for detecting MRD in these solid tumors.
Collapse
Affiliation(s)
- Selena Y Lin
- Department of Translational Molecular Medicine, John Wayne Cancer Institute, Santa Monica, CA, USA
| | - Javier I J Orozco
- Department of Translational Molecular Medicine, John Wayne Cancer Institute, Santa Monica, CA, USA
| | - Dave S B Hoon
- Department of Translational Molecular Medicine, John Wayne Cancer Institute, Santa Monica, CA, USA.
| |
Collapse
|
18
|
Abstract
The field of genomic biomarkers in melanoma has evolved dramatically in the past few decades. Whereas much of the prior focus was on molecular assessment of tumor tissue, circulating tumor cells (CTCs), and cell-free circulating tumor DNA (ctDNA) as sources of a "liquid biopsy" in cancer patients provide promising potential as a method to assess tumor progression, identify targets for therapy, and evaluate clinical response to treatment. Blood biomarker assays have the advantage of being noninvasive, allow for dynamic evaluation of disease over a serial time frame, and help to address the issue of tissue sampling bias and tumor heterogeneity. However, there remains an assortment of technologies and techniques to isolate and detect CTCs and ctDNA and a standardized method has yet to be established. Despite these challenges, multiple studies have already demonstrated the clinical prognostic utility of blood-based genomic biomarker assays. With the advent of next-generation sequencing and genome-wide ctDNA analysis, this will undoubtedly lead to an improved understanding of tumor progression, help to identify new targets for treatment, and improve monitoring of treatment response and development of resistance.
Collapse
Affiliation(s)
- Kelly Huynh
- Department of Surgical Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA 90404
| | - Dave S B Hoon
- Department of Molecular Oncology John Wayne Cancer Institute at Providence Saint John's Health Center Santa Monica, California
| |
Collapse
|
19
|
Autilio C, Paolillo C, Lavieri MM, Pocino K, De Paolis E, Di Stasio E, Marchetti P, Gian Carlo CA, Capoluongo E. PAX3d mRNA over 2.76 copies/µL in the bloodstream predicts cutaneous malignant melanoma relapse. Oncotarget 2017; 8:85479-85491. [PMID: 29156734 PMCID: PMC5689624 DOI: 10.18632/oncotarget.20177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 07/25/2017] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE The aim of this study was to evaluate if our molecular algorithm, based on tumor circulating transcripts, may predict relapse risk in cutaneous malignant melanoma (CMM). RESULTS The multi-marker panel was able to differentiate patients with CMM from HC with high diagnostic sensitivity and specificity, especially for MITF-m and TGFB2 (91-100%) whose levels decreased during follow-up of recurrence-free patients, and remained stable in the case of relapse. PAX3d higher than 2.76 copies/µL emerged as a promising biomarker [specificity = 75-93% and negative predictive value = 75-98%] to stratify subjects at high risk of CMM recurrence independently of age, gender and AJCC staging [OD = 9.5(3.2-28.0), p < 0.001]. The survival analysis confirmed PAX3d performance in relapse prediction with significant differences in recurrence risk 12 months after the basal time-point (p = 0.008). MATERIALS AND METHODS Peripheral blood was collected from 111 CMM patients and from 87 healthy controls (HC) randomly selected. Each specimen was examined by qRT-PCR analysis for the expression of 3 tumor-related transcripts (PAX3d, MITF-m and TGFB2) at diagnosis, and at the following 6 and 12 months during clinical monitoring. CONCLUSIONS We demonstrated the usefulness of our molecular algorithm to indirectly detect circulating melanoma cells in blood, along with PAX3d capability to assess patients' progression and relapse prediction.
Collapse
Affiliation(s)
- Chiara Autilio
- Institute of Clinical Biochemistry, Laboratory of Clinical Molecular Diagnostics, Fondazione Policlinico “A. Gemelli”, Catholic University of the Sacred Heart, Rome, Italy
| | - Carmela Paolillo
- Institute of Clinical Biochemistry, Laboratory of Clinical Molecular Diagnostics, Fondazione Policlinico “A. Gemelli”, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Krizia Pocino
- Institute of Clinical Biochemistry, Laboratory of Clinical Molecular Diagnostics, Fondazione Policlinico “A. Gemelli”, Catholic University of the Sacred Heart, Rome, Italy
| | - Elisa De Paolis
- Institute of Clinical Biochemistry, Laboratory of Clinical Molecular Diagnostics, Fondazione Policlinico “A. Gemelli”, Catholic University of the Sacred Heart, Rome, Italy
| | - Enrico Di Stasio
- Laboratory of Clinical Biochemistry, Fondazione Policlinico “A. Gemelli”, Rome, Italy
| | | | | | - Ettore Capoluongo
- Institute of Clinical Biochemistry, Laboratory of Clinical Molecular Diagnostics, Fondazione Policlinico “A. Gemelli”, Catholic University of the Sacred Heart, Rome, Italy
- Laboratory of Advanced Molecular Diagnostics (DIMA), Istituto Dermopatico dell’Immacolata, Fondazione Luigi Maria Monti, IRCCS, Rome, Italy
- “Molipharma Srl” a Spinoff of Catholic University, Campobasso, Italy
| |
Collapse
|
20
|
Multifunctional, inexpensive, and reusable nanoparticle-printed biochip for cell manipulation and diagnosis. Proc Natl Acad Sci U S A 2017; 114:E1306-E1315. [PMID: 28167769 DOI: 10.1073/pnas.1621318114] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Isolation and characterization of rare cells and molecules from a heterogeneous population is of critical importance in diagnosis of common lethal diseases such as malaria, tuberculosis, HIV, and cancer. For the developing world, point-of-care (POC) diagnostics design must account for limited funds, modest public health infrastructure, and low power availability. To address these challenges, here we integrate microfluidics, electronics, and inkjet printing to build an ultra-low-cost, rapid, and miniaturized lab-on-a-chip (LOC) platform. This platform can perform label-free and rapid single-cell capture, efficient cellular manipulation, rare-cell isolation, selective analytical separation of biological species, sorting, concentration, positioning, enumeration, and characterization. The miniaturized format allows for small sample and reagent volumes. By keeping the electronics separate from microfluidic chips, the former can be reused and device lifetime is extended. Perhaps most notably, the device manufacturing is significantly less expensive, time-consuming, and complex than traditional LOC platforms, requiring only an inkjet printer rather than skilled personnel and clean-room facilities. Production only takes 20 min (vs. up to weeks) and $0.01-an unprecedented cost in clinical diagnostics. The platform works based on intrinsic physical characteristics of biomolecules (e.g., size and polarizability). We demonstrate biomedical applications and verify cell viability in our platform, whose multiplexing and integration of numerous steps and external analyses enhance its application in the clinic, including by nonspecialists. Through its massive cost reduction and usability we anticipate that our platform will enable greater access to diagnostic facilities in developed countries as well as POC diagnostics in resource-poor and developing countries.
Collapse
|
21
|
Adler NR, Haydon A, McLean CA, Kelly JW, Mar VJ. Metastatic pathways in patients with cutaneous melanoma. Pigment Cell Melanoma Res 2016; 30:13-27. [PMID: 27900851 DOI: 10.1111/pcmr.12544] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 10/15/2016] [Indexed: 12/21/2022]
Abstract
Metastasis represents the end product of an elaborate biological process, which is determined by a complex interplay between metastatic tumour cells, host factors and homoeostatic mechanisms. Cutaneous melanoma can metastasize haematogenously or lymphogenously. The three predominant models that endeavour to explain the patterns of melanoma progression are the stepwise spread model, the simultaneous spread model and the model of differential spread. The time course to the development of metastases differs between the different metastatic routes. There are several clinical and histopathological risk factors for the different metastatic pathways. In particular, patient sex and the anatomical location of the primary tumour influence patterns of disease progression. There is limited existing evidence regarding the relationship between tumour mutation status, other diagnostic and prognostic biomarkers and the metastatic pathways of primary cutaneous melanoma. This knowledge gap needs to be addressed to better identify patients at high risk of disease recurrence and personalize surveillance strategies.
Collapse
Affiliation(s)
- Nikki R Adler
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Vic, Australia.,School of Public Health and Preventive Medicine, Monash University, Alfred Hospital, Melbourne, Vic, Australia
| | - Andrew Haydon
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Vic, Australia.,Department of Medical Oncology, Alfred Hospital, Melbourne, Vic, Australia
| | - Catriona A McLean
- Department of Anatomical Pathology, Alfred Hospital, Melbourne, Vic, Australia
| | - John W Kelly
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Vic, Australia
| | - Victoria J Mar
- Victorian Melanoma Service, Alfred Hospital, Melbourne, Vic, Australia.,School of Public Health and Preventive Medicine, Monash University, Alfred Hospital, Melbourne, Vic, Australia.,Skin and Cancer Foundation, Carlton, Vic, Australia
| |
Collapse
|
22
|
Huang SK, Hoon DSB. Liquid biopsy utility for the surveillance of cutaneous malignant melanoma patients. Mol Oncol 2016; 10:450-63. [PMID: 26778792 PMCID: PMC5307330 DOI: 10.1016/j.molonc.2015.12.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 01/01/2023] Open
Abstract
Cutaneous melanoma is one of the highest incident-rate cancers with increasing prevalence in Western societies. Despite the advent of new approved therapeutics, the 5-year overall survival rate of stage IV melanoma patients remains below 15%. Current treatments for late stage disease have shown higher efficacy when treated at a lower disease burden. Thus, blood-based biomarkers capable of detecting melanoma prior to clinically evident distant metastasis, will improve the treatment and outcomes for melanoma patients. To that end, effective treatment of melanoma necessitates identification of patients at risk for developing distant metastases. Furthermore, employing blood biomarkers that monitor cancer progression over the course of treatment is a promising solution to post-treatment drug resistance often developed in melanoma patients. Non-invasive blood biomarker assays allow for regular dynamic monitoring of disease. "Liquid Biopsy" of blood, which exploits circulating tumor cells (CTCs), cell-free circulating tumor DNA (ctDNA) and cell-free circulating microRNA (cmiRNA), has been shown to detect prognostic factors for relapse in AJCC stage III and stage IV melanoma patients. Moreover, molecular characterization of CTC and analysis of various forms of ctDNA present promising potential in development of individualized therapy for melanoma patients. New approaches such as massive parallel sequencing (MPS) provide a comprehensive view of the disease progression, allowing for the selection of therapeutic options for individual patients. With advancements of improving molecular assays, liquid biopsy analysis as a powerful, routine clinical assay for melanoma patients, is highly promising prospective.
Collapse
Affiliation(s)
- Sharon K Huang
- Department of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA 90404, United States
| | - Dave S B Hoon
- Department of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA 90404, United States.
| |
Collapse
|
23
|
Danila DC, Scher HI, Fleisher M. Circulating Tumor Cells as an Analytical Tool in the Management of Patients with Cancer. MANUAL OF MOLECULAR AND CLINICAL LABORATORY IMMUNOLOGY 2016:1051-1061. [DOI: 10.1128/9781555818722.ch111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Xu MJ, Dorsey JF, Amaravadi R, Karakousis G, Simone CB, Xu X, Xu W, Carpenter EL, Schuchter L, Kao GD. Circulating Tumor Cells, DNA, and mRNA: Potential for Clinical Utility in Patients With Melanoma. Oncologist 2015; 21:84-94. [PMID: 26614709 DOI: 10.1634/theoncologist.2015-0207] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/08/2015] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED : Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and messenger RNA (mRNA), collectively termed circulating tumor products (CTPs), represent areas of immense interest from scientists' and clinicians' perspectives. In melanoma, CTP analysis may have clinical utility in many areas, from screening and diagnosis to clinical decision-making aids, as surveillance biomarkers or sources of real-time genetic or molecular characterization. In addition, CTP analysis can be useful in the discovery of new biomarkers, patterns of treatment resistance, and mechanisms of metastasis development. Here, we compare and contrast CTCs, ctDNA, and mRNA, review the extent of translational evidence to date, and discuss how future studies involving both scientists and clinicians can help to further develop this tool for the benefit of melanoma patients. IMPLICATIONS FOR PRACTICE Scientific advancement has enabled the rapid development of tools to analyze circulating tumor cells, tumor DNA, and messenger RNA, collectively termed circulating tumor products (CTPs). A variety of techniques have emerged to detect and characterize melanoma CTPs; however, only a fraction has been applied to human subjects. This review summarizes the available human data that investigate clinical utility of CTP in cancer screening, melanoma diagnosis, prognosis, prediction, and genetic or molecular characterization. It provides a rationale for how CTPs may be useful for future research and discusses how clinicians can be involved in developing this exciting new technology.
Collapse
Affiliation(s)
- Melody J Xu
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jay F Dorsey
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ravi Amaravadi
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Giorgos Karakousis
- Division of Surgical Oncology, Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Charles B Simone
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiaowei Xu
- Department of Pathology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wei Xu
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erica L Carpenter
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lynn Schuchter
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gary D Kao
- Department of Radiation Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Abstract
Circulating melanoma cells (CMCs) represent critical mediators of metastatic melanoma progression. However, isolation and characterization of CMCs has been challenging due to the low frequency of these cells and the paucity of melanoma-specific cell surface markers. Herein, we describe a method for the isolation of CMCs that employs two independent markers, displays high sensitivity for CMC enrichment, and can be readily adapted to include additional molecular melanoma markers of interest. CMCs isolated by this method are enriched for ABCB5-positive melanoma stem cells, are tumorigenic in xenotransplantation assays, and can be used for phenotypical, genetic, and functional investigations of CMC biology.
Collapse
Affiliation(s)
- Jie Ma
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Transplantation Research Program, Division of Nephrology, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Transplantation Research Program, Division of Nephrology, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Roland CL, Ross MI, Hall CS, Laubacher B, Upshaw J, Anderson AE, Lucci A. Detection of circulating melanoma cells in the blood of melanoma patients: a preliminary study. Melanoma Res 2015; 25:335-41. [PMID: 26011119 PMCID: PMC5642955 DOI: 10.1097/cmr.0000000000000168] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significant prognostic heterogeneity exists within the substages of melanoma; therefore, novel prognostic biomarkers are needed to provide information on the risk of recurrence. Limited available data suggest prognostic significance for circulating melanoma cells (CMCs); there is a need for a sensitive, reproducible, and standardized identification technique. Using a semiautomated technology, we sought to determine whether CMCs could be identified reliably in stage I-IV melanoma patients and whether the presence of CMC correlated with known prognostic factors. CMCs were detected in the peripheral blood (7.5 ml) of patients with stage I-IV melanoma (n=89) using the CellSearch system. CD146 cells were immunomagnetically enriched; nucleated HMW-MAA/CD45/CD34 cells were considered CMCs. One or more CMCs was detected in 45% of all patients, varying with stage of disease (stages I/II, III, and IV: 35, 44, and 86%, respectively; P=0.03, for stage I/II vs. stage IV); 55% had one CMC, 32% had two CMCs, and 13% had three or more CMCs identified. The presence of CMCs in the blood was associated with histologic subtype, particularly in patients with stage I/II disease (superficial spreading 18% vs. acral lentiginous 75%). Using a semiautomated technique, CMCs can be identified in a significant number of melanoma patients. These data support further study with longer follow-up and longitudinal/serial time points to better determine the identification rates and prognostic significance of CMCs in stage I-IV melanoma patients.
Collapse
Affiliation(s)
- Christina L Roland
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhang C, Wang L, Guan Y, Sun Y, Liu X, Zhu D, Guo Q. Progress of Circulating Tumor Cells in Cancer Management. Technol Cancer Res Treat 2015; 15:509-16. [PMID: 25948322 DOI: 10.1177/1533034615583762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 03/30/2015] [Indexed: 12/28/2022] Open
Abstract
Circulating tumor cells are low-frequency cells that are shed into the peripheral bloodstream from a primary solid tumor and/or metastasis. Although these cells were recognized initially in 1869, it is only in the past 2 decades that they have been isolated for use as a surrogate biomarker to monitor response to therapy, evaluate prognosis, detect tumor mutations, assist in selecting personalized medicine, and enable earlier cancer diagnosis.
Collapse
Affiliation(s)
- Chufeng Zhang
- School of Medicine and Life Sciences, University of Jinan-Shandong, Academy of Medical Sciences, Jinan, China Academy of Medical Sciences, Shandong Cancer Hospital, Jinan, Shandong, China
| | - Lijie Wang
- School of Medicine and Life Sciences, University of Jinan-Shandong, Academy of Medical Sciences, Jinan, China Academy of Medical Sciences, Shandong Cancer Hospital, Jinan, Shandong, China
| | - Yan Guan
- Academy of Medical Sciences, Shandong Cancer Hospital, Jinan, Shandong, China
| | - Yulan Sun
- Academy of Medical Sciences, Shandong Cancer Hospital, Jinan, Shandong, China
| | - Xiuju Liu
- Academy of Medical Sciences, Shandong Cancer Hospital, Jinan, Shandong, China
| | - Dongyuan Zhu
- Academy of Medical Sciences, Shandong Cancer Hospital, Jinan, Shandong, China
| | - Qisen Guo
- Academy of Medical Sciences, Shandong Cancer Hospital, Jinan, Shandong, China
| |
Collapse
|
28
|
Lau E, Sedy J, Sander C, Shaw MA, Feng Y, Scortegagna M, Claps G, Robinson S, Cheng P, Srivas R, Soonthornvacharin S, Ideker T, Bosenberg M, Gonzalez R, Robinson W, Chanda SK, Ware C, Dummer R, Hoon D, Kirkwood JM, Ronai ZA. Transcriptional repression of IFNβ1 by ATF2 confers melanoma resistance to therapy. Oncogene 2015; 34:5739-48. [PMID: 25728676 PMCID: PMC4558399 DOI: 10.1038/onc.2015.22] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/02/2015] [Accepted: 01/06/2015] [Indexed: 02/07/2023]
Abstract
The resistance of melanoma to current treatment modalities represents a major obstacle for durable therapeutic response, and thus, the elucidation of mechanisms of resistance is urgently needed. The crucial functions of Activating Transcription Factor-2 (ATF2) in the development and therapeutic resistance of melanoma have been previously reported, although the precise underlying mechanisms remain unclear. Here, we report a protein kinase C epsilon (PKCε)- and Activating Transcription Factor-2 (ATF2)-mediated mechanism that facilitates resistance by transcriptionally repressing the expression of IFNβ1 and downstream type-I IFN signaling, which is otherwise induced upon exposure to chemotherapy. Treatment of early stage melanomas expressing low levels of PKCε with chemotherapies relieves its transcriptional repression of IFNB1, resulting in impaired S-phase progression, a senescence-like phenotype, and increased cell death. This response is lost in late stage metastatic melanomas expressing high levels of PKCε. Notably, nuclear ATF2 and low expression of IFNβ1 in melanoma tumor samples correlates with poor patient responsiveness to biochemotherapy or neoadjuvant IFN-α2a. Conversely, cytosolic ATF2 and induction of IFNβ1 coincides with therapeutic responsiveness. Collectively, we identify an IFNβ1-dependent, cell autonomous mechanism that contributes to the therapeutic resistance of melanoma via the PKCε-ATF2 regulatory axis.
Collapse
Affiliation(s)
- E Lau
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - J Sedy
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - C Sander
- University of Pittsburgh Cancer Center, Pittsburgh, PA, USA
| | - M A Shaw
- John Wayne Cancer Institute, Santa Monica, CA, USA
| | - Y Feng
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - M Scortegagna
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - G Claps
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - S Robinson
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - P Cheng
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - R Srivas
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - S Soonthornvacharin
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - T Ideker
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | | | - R Gonzalez
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - W Robinson
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - S K Chanda
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - C Ware
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - R Dummer
- Department of Dermatology, University of Zurich, Zurich, Switzerland
| | - D Hoon
- John Wayne Cancer Institute, Santa Monica, CA, USA
| | - J M Kirkwood
- University of Pittsburgh Cancer Center, Pittsburgh, PA, USA
| | - Z A Ronai
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
29
|
Marzese DM, Liu M, Huynh JL, Hirose H, Donovan NC, Huynh KT, Kiyohara E, Chong K, Cheng D, Tanaka R, Morton DL, Barkhoudarian G, Kelly DF, Hoon DS. Brain metastasis is predetermined in early stages of cutaneous melanoma by CD44v6 expression through epigenetic regulation of the spliceosome. Pigment Cell Melanoma Res 2015; 28:82-93. [PMID: 25169209 PMCID: PMC4309554 DOI: 10.1111/pcmr.12307] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 08/12/2014] [Accepted: 08/12/2014] [Indexed: 12/20/2022]
Abstract
Melanoma brain metastasis (MBM) is frequent and has a very poor prognosis with no current predictive factors or therapeutic molecular targets. Our study unravels the molecular alterations of cell-surface glycoprotein CD44 variants during melanoma progression to MBM. High expression of CD44 splicing variant 6 (CD44v6) in primary melanoma (PRM) and regional lymph node metastases from AJCC Stage IIIC patients significantly predicts MBM development. The expression of CD44v6 also enhances the migration of MBM cells by hyaluronic acid and hepatocyte growth factor exposure. Additionally, CD44v6-positive MBM migration is reduced by blocking with a CD44v6-specific monoclonal antibody or knocking down CD44v6 by siRNA. ESRP1 and ESRP2 splicing factors correlate with CD44v6 expression in PRM, and ESRP1 knockdown significantly decreases CD44v6 expression. However, an epigenetic silencing of ESRP1 is observed in metastatic melanoma, specifically in MBM. In advanced melanomas, CD44v6 expression correlates with PTBP1 and U2AF2 splicing factors, and PTBP1 knockdown significantly decreases CD44v6 expression. Overall, these findings open a new avenue for understanding the high affinity of melanoma to progress to MBM, suggesting CD44v6 as a potential MBM-specific factor with theranostic utility for stratifying patients.
Collapse
Affiliation(s)
- Diego M. Marzese
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Michelle Liu
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Jamie L. Huynh
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Hajime Hirose
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Nicholas C. Donovan
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Kelly T. Huynh
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Eiji Kiyohara
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Kelly Chong
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - David Cheng
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Ryo Tanaka
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Donald L. Morton
- Division of Surgical Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Garni Barkhoudarian
- Brain Tumor Center & Pituitary Disorders Program, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Daniel F. Kelly
- Brain Tumor Center & Pituitary Disorders Program, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| | - Dave S.B. Hoon
- Dept of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, CA 90404, USA
| |
Collapse
|
30
|
Abstract
Treatment strategies for metastatic melanoma have changed markedly in recent times due to the advent of targeted systemic therapies. In addition to these developments, surgery remains a useful adjunct that can confer survival benefits in selected patients. In this review, we examine the current literature to highlight the role of surgical intervention in metastatic melanoma in the era of targeted systemic therapies.
Collapse
|
31
|
Abstract
Melanoma has traditionally been associated with limited treatment options, and as such, biomarkers such as histopathologic staging and serum lactate dehydrogenase focused on prognosis. The development of effective treatment options shifted the search to biomarkers for predicting response and resistance to therapy, an arguably more critical goal. Specific genetic alterations (e.g., BRAFV600 and KIT mutations) predict response to molecularly targeted agents and are routinely used in clinical practice. Other promising biomarkers include T-cell characteristics (the circulating and tumor microenvironment), tumor expression of PD-L1, circulating DNA, circulating tumor cells and miRNAs. In this article, we discuss the status of the currently used and experimental tumor- and blood-based biomarkers for melanoma prognosis and response to targeted and immune therapies.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research building, Nashville, TN 37232, USA.,Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research building, Nashville, TN 37232, USA
| | - Ryan J Sullivan
- Department of Medicine, Division of Hematology/Oncology Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.,Department of Medicine, Division of Hematology/Oncology Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
32
|
Leong SPL, Tseng WW. Micrometastatic cancer cells in lymph nodes, bone marrow, and blood: Clinical significance and biologic implications. CA Cancer J Clin 2014; 64:195-206. [PMID: 24500995 DOI: 10.3322/caac.21217] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 01/09/2023] Open
Abstract
Cancer metastasis may be regarded as a progressive process from its inception in the primary tumor microenvironment to distant sites by way of the lymphovascular system. Although this type of tumor dissemination often occurs in an orderly fashion via the sentinel lymph node (SLN), acting as a possible gateway to the regional lymph nodes, bone marrow, and peripheral blood and ultimately to distant metastatic sites, this is not a general rule as tumor cells may enter the blood and spread to distant sites, bypassing the SLN. Methods of detecting micrometastatic cancer cells in the SLN, bone marrow, and peripheral blood of patients have been established. Patients with cancer cells in their SLN, bone marrow, or peripheral blood have worse clinical outcomes than patients with no evidence of spread to these compartments. The presence of these cells also has important biologic implications for disease progression and the clinician's understanding of the process of cancer metastasis. Further characterization of these micrometastatic cancer cells at each stage and site of metastasis is needed to design novel selective therapies for a more "personalized" treatment.
Collapse
Affiliation(s)
- Stanley P L Leong
- Chief of Cutaneous Oncology, Associate Director of the Melanoma Program, Center for Melanoma Research and Treatment, California Pacific Medical Center and Sutter Pacific Medical Foundation, Senior Scientist, California Pacific Medical Center Research Institute, San Francisco, CA
| | | |
Collapse
|
33
|
Shtivelman E, Davies MA, Hwu P, Yang J, Lotem M, Oren M, Flaherty KT, Fisher DE. Pathways and therapeutic targets in melanoma. Oncotarget 2014; 5:1701-52. [PMID: 24743024 PMCID: PMC4039128 DOI: 10.18632/oncotarget.1892] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 02/07/2023] Open
Abstract
This review aims to summarize the current knowledge of molecular pathways and their clinical relevance in melanoma. Metastatic melanoma was a grim diagnosis, but in recent years tremendous advances have been made in treatments. Chemotherapy provided little benefit in these patients, but development of targeted and new immune approaches made radical changes in prognosis. This would not have happened without remarkable advances in understanding the biology of disease and tremendous progress in the genomic (and other "omics") scale analyses of tumors. The big problems facing the field are no longer focused exclusively on the development of new treatment modalities, though this is a very busy area of clinical research. The focus shifted now to understanding and overcoming resistance to targeted therapies, and understanding the underlying causes of the heterogeneous responses to immune therapy.
Collapse
Affiliation(s)
| | | | - Patrick Hwu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James Yang
- National Cancer Institute, NIH, Washington DC, USA
| | - Michal Lotem
- Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Moshe Oren
- The Weizmann Institute of Science, Rehovot, Israel
| | | | - David E. Fisher
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
34
|
Chiu CG, Nakamura Y, Chong KK, Huang SK, Kawas NP, Triche T, Elashoff D, Kiyohara E, Irie RF, Morton DL, Hoon DSB. Genome-wide characterization of circulating tumor cells identifies novel prognostic genomic alterations in systemic melanoma metastasis. Clin Chem 2014; 60:873-85. [PMID: 24718909 DOI: 10.1373/clinchem.2013.213611] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Circulating tumor cells (CTC) have been found in patients with metastatic melanoma and are associated with advanced melanoma stage and poor patient outcome. We hypothesize that CTC harbor genomic changes critical in the development of distant systemic metastasis. Here, we present the first genome-wide copy-number aberration (CNA) and loss of heterozygosity (LOH)-based characterization of melanoma CTC. METHODS CTC were isolated from peripheral blood monocytes of 13 melanoma patients with regional metastasis stage IIIB/C using antibodies against melanoma-associated cell surface gangliosides. RESULTS We characterized 251 CNA in CTC. Comparative analysis demonstrated >90% concordance in single-nucleotide polymorphism profiles between paired CTC and tumor metastases. In particular, there were notable recurring CNA across patients. In exploratory studies, the presence of several top CTC-associated CNA was verified in distant metastasis (stage IV) from 27 patients, suggesting that certain genomic changes are propagated from regional metastasis to CTC and to distant systemic metastases. Lastly, an exploratory biomarker panel derived from 5 CTC-associated CNA [CSMD2 (CUB and Sushi multiple domains 2), 1p35.1; CNTNAP5 (contactin associated protein-like 5), 2q14.3; NRDE2 (NRDE-2, necessary for RNA interference, domain containing), 14q32.11; ADAM6 (ADAM metallopeptidase domain 6, pseudogene), 14q32.33; and TRPM2 (transient receptor potential cation channel, subfamily m, member 2), 21q22.3] conferred prognostic utility for melanoma recurrence [hazard ratio (HR), 1.14; CI, 1.00-1.44; P = 0.0471] and death (HR, 2.86; CI, 1.23-14.42; P = 0.0014) in 35 patients with stage IIIB/C melanoma, with a 5-year disease-free survival of 13% vs 69% (P = 0.0006) and overall survival of 28% vs 94% between high-risk and low-risk groups defined by the biomarker panel, respectively. CONCLUSIONS This study provides the first detailed CNA-based profile of melanoma CTC and illustrates how CTC may be used as a novel approach for identification of systemic metastasis.
Collapse
Affiliation(s)
- Connie G Chiu
- Department of Molecular Oncology and Division of Surgical Oncology, John Wayne Cancer Institute, Santa Monica, CA
| | | | | | | | | | - Timothy Triche
- Center for Personalized Medicine, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David Elashoff
- Department of Medicine Statistics Core, UCLA School of Medicine, Los Angeles, CA
| | | | - Reiko F Irie
- Department of Biotechnology, John Wayne Cancer Institute, Santa Monica, CA
| | - Donald L Morton
- Division of Surgical Oncology, John Wayne Cancer Institute, Santa Monica, CA
| | | |
Collapse
|
35
|
Rodic S, Mihalcioiu C, Saleh RR. Detection methods of circulating tumor cells in cutaneous melanoma: a systematic review. Crit Rev Oncol Hematol 2014; 91:74-92. [PMID: 24530125 DOI: 10.1016/j.critrevonc.2014.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/06/2014] [Accepted: 01/14/2014] [Indexed: 12/25/2022] Open
Abstract
The vast majority of melanoma-related deaths are due to disseminated malignancy. Many treated patients who are clinically disease-free will go on to relapse. Therefore, new prognostic tools must be developed to better assess metastatic potential and assist in patient management. Circulating tumor cells are a widely studied metastatic biomarker with promising prognostic utility, as the shedding of cells from the primary tumor into peripheral blood is a necessary step in disease dissemination. An assortment of technologies and techniques has been developed to isolate and detect circulating melanoma cells (CMCs), but a standardized method is yet to be established. It is the aim of this study to systematically review the diverse enrichment and detection methods of circulating tumor cells in cutaneous melanoma. A literature search yielded 351 articles, of which 74 were deemed eligible according to inclusion criteria, the primary requirement being the reporting of patient CMC positivity status stratified by the stage of melanoma. Pertinent studies were used to evaluate the advantages and disadvantages of each method. Additionally, we calculated the sensitivity and specificity of seven common melanoma-associated markers based on the available literature.
Collapse
Affiliation(s)
- Stefan Rodic
- Division of Biology, McGill University, Montreal, Canada
| | - Catalin Mihalcioiu
- Division of Medical Oncology, McGill University Health Centre, Montreal, Canada
| | - Ramy R Saleh
- Division of Medical Oncology, McGill University Health Centre, Montreal, Canada.
| |
Collapse
|
36
|
Itakura E, Cochran A. Diagnostic and Prognostic Biomarkers in Cutaneous Melanoma. Cancer Biomark 2014. [DOI: 10.1201/b16389-41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Kiyohara E, Hata K, Lam S, Hoon DSB. Circulating tumor cells as prognostic biomarkers in cutaneous melanoma patients. Methods Mol Biol 2014; 1102:513-22. [PMID: 24258996 DOI: 10.1007/978-1-62703-727-3_27] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Detection of circulating tumor cells (CTC) in peripheral blood has been investigated for its prognostic ability, and its potential to measure the effectiveness of treatment(s) in patients with melanoma. However, a highly sensitive and specific assay is required to detect CTC in patients' blood. We have developed a multimarker quantitative real-time reverse transcriptase polymerase chain reaction (RT-qPCR) assay for detecting CTC directly from peripheral blood specimens without the need of separating CTC from leukocytes (PBL). We selected and optimized four mRNA biomarkers (MART-1/Melan-A, MAGE-A3, PAX3, and GalNAc-T) for detection and prediction of clinical outcome in melanoma patients. Our protocol has both high sensitivity and specificity for CTC in blood specimens-detecting approximately one to five melanoma cells in 10(7) PBL. We have demonstrated the significance of this assay for serial bleed assessment of CTC in clinical trials and for daily clinical usage.
Collapse
Affiliation(s)
- Eiji Kiyohara
- Department of Molecular Oncology, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica, CA, USA
| | | | | | | |
Collapse
|
38
|
Blom A, Bhatia S, Pietromonaco S, Koehler K, Iyer JG, Nagase K, Paulson K, Sabath DE, Nghiem P. Clinical utility of a circulating tumor cell assay in Merkel cell carcinoma. J Am Acad Dermatol 2013; 70:449-55. [PMID: 24388423 DOI: 10.1016/j.jaad.2013.10.051] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 10/07/2013] [Accepted: 10/28/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND Quantitation of circulating tumor cells (CTCs) has utility in managing breast, colon, and prostate carcinomas. OBJECTIVE We sought to determine whether a commercially available CTC assay provides prognostic information in Merkel cell carcinoma (MCC), insight into treatment responses, or both. METHODS We analyzed CTCs in 52 specimens from 34 patients with MCC. RESULTS The presence of CTCs correlated with extent of disease at blood draw (P = .004). Among 15 patients with regional nodal disease, CTC-negative patients had 80% disease-specific survival at 2 years after the test, versus 29% for CTC-positive patients (P = .015). Among the entire cohort, those without CTCs had 72% MCC-specific survival whereas CTC-positive patients had 25% survival (n = 34, median follow-up 19 months, P = .0003). Fifty seven percent of patients with MCC had a cytokeratin "dot" visible in 20% or more of CTCs, a feature that was absent among CTCs from other carcinomas (0 of 13 cases). LIMITATIONS CTC assay was performed at variable times after diagnosis and heterogeneity in extent of disease affects interpretability of the data. CONCLUSION CTC detection in MCC is feasible and appears to add prognostic information, particularly in patients with regional nodal disease. It may also assist clinical management in certain situations, including differentiating metastatic MCC cells from those of other carcinomas.
Collapse
Affiliation(s)
- Astrid Blom
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Shailender Bhatia
- Department of Medicine/Oncology, University of Washington, Seattle, Washington
| | | | - Karen Koehler
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Jayasri G Iyer
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Kotaro Nagase
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Kelly Paulson
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Daniel E Sabath
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Paul Nghiem
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington.
| |
Collapse
|
39
|
Marzese DM, Hirose H, Hoon DSB. Diagnostic and prognostic value of circulating tumor-related DNA in cancer patients. Expert Rev Mol Diagn 2013; 13:827-844. [PMID: 24127721 DOI: 10.1586/14737159.2013.845088] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Qualitative and quantitative analysis of circulating cell-free DNA (cfDNA) is an emerging non-invasive blood biomarker utilized to assess tumor progression and to evaluate prognosis, diagnosis and response to treatment. There is a need to develop cfDNA biomarkers to avoid complex risk-prone biopsy procedures for primary or metastatic tumors. Given the challenges associated with inter- and intra-tumor heterogeneity, the implementation of genome-wide cfDNA analysis will become an important avenue to understand tumor progression and therapeutic settings, not only for predominant, but also for under-represented tumor subclones with specific genomic aberrations. We summarize the latest publications in cfDNA analysis, including a metric analysis of clinical trials and new high-throughput technology applied to cfDNA analysis in clinical oncology.
Collapse
Affiliation(s)
- Diego M Marzese
- Department of Molecular Oncology, John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA, 90404, USA
| | | | | |
Collapse
|
40
|
Joshi P, Zborowski M, Triozzi PL. Circulating melanoma cells: scoping the target. Front Oncol 2013; 3:189. [PMID: 23964346 PMCID: PMC3740478 DOI: 10.3389/fonc.2013.00189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/07/2013] [Indexed: 01/05/2023] Open
Affiliation(s)
- Powrnima Joshi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic Cleveland, OH, USA
| | | | | |
Collapse
|
41
|
Klinac D, Gray ES, Millward M, Ziman M. Advances in personalized targeted treatment of metastatic melanoma and non-invasive tumor monitoring. Front Oncol 2013; 3:54. [PMID: 23515890 PMCID: PMC3601325 DOI: 10.3389/fonc.2013.00054] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/28/2013] [Indexed: 12/19/2022] Open
Abstract
Despite extensive scientific progress in the melanoma field, treatment of advanced stage melanoma with chemotherapeutics and biotherapeutics has rarely provided response rates higher than 20%. In the past decade, targeted inhibitors have been developed for metastatic melanoma, leading to the advent of more personalized therapies of genetically characterized tumors. Here we review current melanoma treatments and emerging targeted molecular therapies. In particular we discuss the mutant BRAF inhibitors Vemurafenib and Dabrafenib, which markedly inhibit tumor growth and advance patients' overall survival. However this response is almost inevitably followed by complete tumor relapse due to drug resistance hampering the encouraging initial responses. Several mechanisms of resistance within and outside the MAPK pathway have now been uncovered and have paved the way for clinical trials of combination therapies to try and overcome tumor relapse. It is apparent that personalized treatment management will be required in this new era of targeted treatment. Circulating tumor cells (CTCs) provide an easily accessible means of monitoring patient relapse and several new approaches are available for the molecular characterization of CTCs. Thus CTCs provide a monitoring tool to evaluate treatment efficacy and early detection of drug resistance in real time. We detail here how advances in the molecular analysis of CTCs may provide insight into new avenues of approaching therapeutic options that would benefit personalized melanoma management.
Collapse
Affiliation(s)
- Dragana Klinac
- School of Medical Sciences, Edith Cowan UniversityPerth, WA, Australia
| | - Elin S. Gray
- School of Medical Sciences, Edith Cowan UniversityPerth, WA, Australia
| | - Michael Millward
- School of Medicine and Pharmacology, University of Western AustraliaCrawley, WA, Australia
| | - Mel Ziman
- School of Medical Sciences, Edith Cowan UniversityPerth, WA, Australia
- School of Pathology and Laboratory Medicine, University of Western AustraliaCrawley, WA, Australia
| |
Collapse
|
42
|
Detection and quantification of CSF malignant cells by the CellSearch® technology in patients with melanoma leptomeningeal metastasis. Med Oncol 2013; 30:538. [DOI: 10.1007/s12032-013-0538-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 03/06/2013] [Indexed: 12/23/2022]
|
43
|
Shim S, Stemke-Hale K, Noshari J, Becker FF, Gascoyne PRC. Dielectrophoresis has broad applicability to marker-free isolation of tumor cells from blood by microfluidic systems. BIOMICROFLUIDICS 2013; 7:11808. [PMID: 24403990 PMCID: PMC3562275 DOI: 10.1063/1.4774307] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 11/21/2012] [Indexed: 05/07/2023]
Abstract
The number of circulating tumor cells (CTCs) found in blood is known to be a prognostic marker for recurrence of primary tumors, however, most current methods for isolating CTCs rely on cell surface markers that are not universally expressed by CTCs. Dielectrophoresis (DEP) can discriminate and manipulate cancer cells in microfluidic systems and has been proposed as a molecular marker-independent approach for isolating CTCs from blood. To investigate the potential applicability of DEP to different cancer types, the dielectric and density properties of the NCI-60 panel of tumor cell types have been measured by dielectrophoretic field-flow fractionation (DEP-FFF) and compared with like properties of the subpopulations of normal peripheral blood cells. We show that all of the NCI-60 cell types, regardless of tissue of origin, exhibit dielectric properties that facilitate their isolation from blood by DEP. Cell types derived from solid tumors that grew in adherent cultures exhibited dielectric properties that were strikingly different from those of peripheral blood cell subpopulations while leukemia-derived lines that grew in non-adherent cultures exhibited dielectric properties that were closer to those of peripheral blood cell types. Our results suggest that DEP methods have wide applicability for the surface-marker independent isolation of viable CTCs from blood as well as for the concentration of leukemia cells from blood.
Collapse
Affiliation(s)
- Sangjo Shim
- Department of Imaging Physics Research, The University of Texas, M.D. Anderson Cancer Center Unit 951, 1515 Holcombe Boulevard, Houston, Texas 77030, USA ; Department of Biomedical Engineering, The University of Texas at Austin, 1 University Station, C0800, Austin, Texas 78712, USA
| | - Katherine Stemke-Hale
- Department of Systems Biology, The University of Texas, M.D. Anderson Cancer Center Unit 951, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Jamileh Noshari
- Department of Imaging Physics Research, The University of Texas, M.D. Anderson Cancer Center Unit 951, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Frederick F Becker
- Department of Molecular Pathology, The University of Texas, M.D. Anderson Cancer Center Unit 951, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Peter R C Gascoyne
- Department of Imaging Physics Research, The University of Texas, M.D. Anderson Cancer Center Unit 951, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| |
Collapse
|
44
|
Reid AL, Millward M, Pearce R, Lee M, Frank MH, Ireland A, Monshizadeh L, Rai T, Heenan P, Medic S, Kumarasinghe P, Ziman M. Markers of circulating tumour cells in the peripheral blood of patients with melanoma correlate with disease recurrence and progression. Br J Dermatol 2012; 168:85-92. [PMID: 23013138 DOI: 10.1111/bjd.12057] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Multimarker quantitative real-time polymerase chain reaction (qRT-PCR) represents an effective method for detecting circulating tumour cells in the peripheral blood of patients with melanoma. OBJECTIVES To investigate whether the phenotype of circulating melanoma cells represents a useful indicator of disease stage, recurrence and treatment efficacy. METHODS Peripheral blood was collected from 230 patients with melanoma and 152 healthy controls over a period of 3years and 9months. Clinical data and blood samples were collected from patients with primary melanoma (early stages, 0-II, n=154) and metastatic melanoma (late stages, III-IV, n=76). Each specimen was examined by qRT-PCR analysis for the expression of five markers: MLANA, ABCB5, TGFβ2, PAX3d and MCAM. RESULTS In total, 212 of the patients with melanoma (92%) expressed markers in their peripheral blood. Two markers, MLANA and ABCB5, had the greatest prognostic value, and were identified as statistically significant among patients who experienced disease recurrence within our study period, being expressed in 45% (MLANA) and 49% (ABCB5) of patients with recurrence (P=0·001 and P=0·031, respectively). For patients administered nonsurgical treatments, MCAM expression correlated with poor treatment outcome. CONCLUSIONS Circulating tumour cells were detectable at all stages of disease and long after surgical treatment, even when patients were considered disease free. Specifically, expression of ABCB5 and MLANA had significant prognostic value in inferring disease recurrence, while MCAM expression was associated with poor patient outcome after treatment, confirming multimarker qRT-PCR as a potential technique for monitoring disease status.
Collapse
Affiliation(s)
- A L Reid
- School of Medical Sciences, Edith Cowan University, Perth, WA, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hoshimoto S, Shingai T, Morton DL, Kuo C, Faries MB, Chong K, Elashoff D, Wang HJ, Elashoff RM, Hoon DS. 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; 30:3819-26. [PMID: 23008288 PMCID: PMC3478576 DOI: 10.1200/jco.2011.40.0887] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 07/24/2012] [Indexed: 01/27/2023] Open
Abstract
PURPOSE The outcomes of patients with melanoma who have sentinel lymph node (SLN) metastases can be highly variable, which has precluded establishment of consensus regarding treatment of the group. The detection of high-risk patients from this clinical setting may be helpful for determination of both prognosis and management. We report the utility of multimarker reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) detection of circulating tumor cells (CTCs) in patients with melanoma diagnosed with SLN metastases in a phase III, international, multicenter clinical trial. PATIENTS AND METHODS Blood specimens were collected from patients with melanoma (n = 331) who were clinically disease-free after complete lymphadenectomy (CLND) before entering onto a randomized adjuvant melanoma vaccine plus bacillus Calmette-Guérin (BCG) versus BCG placebo trial from 30 melanoma centers (United States and international). Blood was assessed using a verified multimarker RT-qPCR assay (MART-1, MAGE-A3, and GalNAc-T) of melanoma-associated proteins. Cox regression analyses were used to evaluate the prognostic significance of CTC status for disease recurrence and melanoma-specific survival (MSS). RESULTS Individual CTC biomarker detection ranged from 13.4% to 17.5%. There was no association of CTC status (zero to one positive biomarkers v two or more positive biomarkers) with known clinical or pathologic prognostic variables. However, two or more positive biomarkers was significantly associated with worse distant metastasis disease-free survival (hazard ratio [HR] = 2.13, P = .009) and reduced recurrence-free survival (HR = 1.70, P = .046) and MSS (HR = 1.88, P = .043) in a multivariable analysis. CONCLUSION CTC biomarker status is a prognostic factor for recurrence-free survival, distant metastasis disease-free survival, and MSS after CLND in patients with SLN metastasis. This multimarker RT-qPCR analysis may therefore be useful in discriminating patients who may benefit from aggressive adjuvant therapy or stratifying patients for adjuvant clinical trials.
Collapse
Affiliation(s)
- Sojun Hoshimoto
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Tatsushi Shingai
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Donald L. Morton
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Christine Kuo
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Mark B. Faries
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Kelly Chong
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - David Elashoff
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - He-Jing Wang
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Robert M. Elashoff
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Dave S.B. Hoon
- Sojun Hoshimoto, Tatsushi Shingai, Donald L. Morton, Christine Kuo, Mark B. Faries, Kelly Chong, and Dave S.B. Hoon, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica; and David Elashoff, He-Jing Wang, and Robert M. Elashoff, University of California, Los Angeles School of Medicine, Los Angeles, CA
| |
Collapse
|
46
|
Abstract
Personalized cancer medicine requires the development of tumor-specific biomarkers to optimize selection of targeted therapies and to better assess response to therapy. Current efforts in several tumor types have shown that patients in whom circulating tumor cells (CTCs) are detected have an inferior prognosis relative to those in whom CTCs are not detected and that the elimination or decrease of CTCs following treatment is associated with improved clinical outcomes. Technological advances in the detection, isolation, capture, and characterization of CTCs from phlebotomy samples obtained in a routine clinical practice setting have enabled the evaluation of different CTC biomarkers. Unmet needs in cancer diagnosis and treatment where CTC biomarkers have been studied include determining prognosis, assessing the effects of treatment, and as a source of tumor for the biologic identification and characterization of determinants to predict sensitivity to one form of treatment versus another and to understand mechanisms of treatment resistance.At present, there is no single definition of a CTC and no single CTC "biomarker." Rather, multiple assays (tests) are in development for CTC biomarkers. However, before the role of any biomarker in medical decision making can be determined, it is essential that the assays used to measure the biomarker are analytically validated in a sequence of trials to generate the evidence to support the biomarker's use in the given context of use. It is against this background that this review focuses on the process of developing CTC biomarker assays, with the objective of outlining the necessary steps to qualify specific CTC tests for medical decision making in clinical practice or drug development. The potential for point-of-care tests is clear.
Collapse
|
47
|
Howard JH, Thompson JF, Mozzillo N, Nieweg OE, Hoekstra HJ, Roses DF, Sondak VK, Reintgen DS, Kashani-Sabet M, Karakousis CP, Coventry BJ, Kraybill WG, Smithers BM, Elashoff R, Stern SL, Cochran AJ, Faries MB, Morton DL. Metastasectomy for distant metastatic melanoma: analysis of data from the first Multicenter Selective Lymphadenectomy Trial (MSLT-I). Ann Surg Oncol 2012; 19:2547-55. [PMID: 22648554 DOI: 10.1245/s10434-012-2398-z] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Indexed: 01/22/2023]
Abstract
BACKGROUND For stage IV melanoma, systemic medical therapy (SMT) is used most frequently; surgery is considered an adjunct in selected patients. We retrospectively compared survival after surgery with or without SMT versus SMT alone for melanoma patients developing distant metastases while enrolled in the first Multicenter Selective Lymphadenectomy Trial. METHODS Patients were randomized to wide excision and sentinel node biopsy, or wide excision and nodal observation. We evaluated recurrence site, therapy (selected by treating clinician), and survival after stage IV diagnosis. RESULTS Of 291 patients with complete data for stage IV recurrence, 161 (55 %) underwent surgery with or without SMT. Median survival was 15.8 versus 6.9 months, and 4-year survival was 20.8 versus 7.0 % for patients receiving surgery with or without SMT versus SMT alone (p < 0.0001; hazard ratio 0.406). Surgery with or without SMT conferred a survival advantage for patients with M1a (median > 60 months vs. 12.4 months; 4-year survival 69.3 % vs. 0; p = 0.0106), M1b (median 17.9 vs. 9.1 months; 4-year survival 24.1 vs. 14.3 %; p = 0.1143), and M1c (median 15.0 vs. 6.3 months; 4-year survival 10.5 vs. 4.6 %; p = 0.0001) disease. Patients with multiple metastases treated surgically had a survival advantage, and number of operations did not reduce survival in the 67 patients (42 %) who had multiple surgeries for distant melanoma. CONCLUSIONS Our findings suggest that over half of stage IV patients are candidates for resection and exhibit improved survival over patients receiving SMT alone, regardless of site and number of metastases. We have begun a multicenter randomized phase III trial comparing surgery versus SMT as initial treatment for resectable distant melanoma.
Collapse
Affiliation(s)
- J Harrison Howard
- Department of Surgical Oncology, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Guida M, Pisconti S, Colucci G. Metastatic melanoma: the new era of targeted therapy. Expert Opin Ther Targets 2012; 16 Suppl 2:S61-70. [DOI: 10.1517/14728222.2011.645807] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Assessment of prognostic circulating tumor cells in a phase III trial of adjuvant immunotherapy after complete resection of stage IV melanoma. Ann Surg 2012; 255:357-62. [PMID: 22202581 DOI: 10.1097/sla.0b013e3182380f56] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE To verify circulating tumor cell (CTC) prognostic utility in stage IV resected melanoma patients in a prospective international phase III clinical trial. BACKGROUND Our studies of melanoma patients in phase II clinical trials demonstrated prognostic significance for CTCs in patients with AJCC stage IV melanoma. CTCs were assessed to determine prognostic utility in follow-up of disease-free stage IV patients pre- and during treatment. METHODS After complete metastasectomy, patients were prospectively enrolled in a randomized trial of adjuvant therapy with a whole-cell melanoma vaccine, Canvaxin, plus Bacille Calmette-Guerin (BCG) versus placebo plus BCG. Blood specimens obtained pretreatment (n = 244) and during treatment (n = 214) were evaluated by quantitative real-time reverse-transcriptase polymerase chain reaction (qPCR) for expression of MART-1, MAGE-A3, and PAX3 mRNA biomarkers. Univariate and multivariate Cox analyses examined CTC biomarker expression with respect to clinicopathological variables. RESULTS CTC biomarker(s) (≥ 1) was detected in 54% of patients pretreatment and in 86% of patients over the first 3 months. With a median follow-up of 21.9 months, 71% of patients recurred and 48% expired. CTC levels were not associated with known prognostic factors or treatment arm. In multivariate analysis, pretreatment CTC (> 0 vs. 0 biomarker) status was significantly associated with disease-free survival (DFS; HR 1.64, P = 0.002) and overall survival (OS; HR 1.53, P = 0.028). Serial CTC (>0 vs. 0 biomarker) status was also significantly associated with DFS (HR 1.91, P = 0.02) and OS (HR 2.57, P = 0.012). CONCLUSION CTC assessment can provide prognostic discrimination before and during adjuvant treatment for resected stage IV melanoma patients.
Collapse
|
50
|
Hoshimoto S, Kuo CT, Chong KK, Takeshima TL, Takei Y, Li MW, Huang SK, Sim MS, Morton DL, Hoon DSB. AIM1 and LINE-1 epigenetic aberrations in tumor and serum relate to melanoma progression and disease outcome. J Invest Dermatol 2012; 132:1689-97. [PMID: 22402438 PMCID: PMC3352986 DOI: 10.1038/jid.2012.36] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aberrations in the methylation status of non-coding genomic repeat DNA sequences and specific gene promoter region are important epigenetic events in melanoma progression. Promoter methylation status in LINE-1 and Absent in melanoma-1(AIM1;6q21) associated with melanoma progression and disease outcome was assessed. LINE-1 and AIM1 methylation status was assessed in paraffin-embedded archival tissues(PEAT)(n=133) and melanoma patients’ serum(n=56). LINE-1 U-Index(hypomethylation) and AIM1 were analyzed in microdissected melanoma PEAT sections. The LINE-1 U-Index of melanoma(n=100) was significantly higher than that of normal skin(n=14) and nevi(n=12)(P=0.0004). LINE-1 U-Index level was elevated with increasing AJCC stage(P<0.0001). AIM1 promoter hypermethylation was found in higher frequency(P=0.005) in metastatic melanoma(65%) than in primary melanomas(38%). When analyzed, high LINE-1 U-Index and/or AIM1 methylation in melanomas were associated with disease-free survival(DFS) and overall survival(OS) in Stage I/II patients (P=0.017, 0.027; respectively). In multivariate analysis, melanoma AIM1 methylation status was a significant prognostic factor of OS(P=0.032). Furthermore, serum unmethylated LINE-1 was at higher levels in both stage III(n=20) and stage IV(n=36) patients compared to healthy donors(n=14)(P=0.022). Circulating methylated AIM1 was detected in patients’ serum and was predictive of OS in Stage IV patients (P=0.009). LINE-1 hypomethylation and AIM1 hypermethylation have prognostic utility in both melanoma patients’ tumors and serum.
Collapse
Affiliation(s)
- Sojun Hoshimoto
- Department of Molecular Oncology, John Wayne Cancer Institute at Saint John's Health Center, Santa Monica, California 90404, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|